[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (21)

Search Parameters:
Keywords = atrogens

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 12312 KiB  
Article
Chikungunya and Mayaro Viruses Induce Chronic Skeletal Muscle Atrophy Triggered by Pro-Inflammatory and Oxidative Response
by Mariana Oliveira Lopes da Silva, Camila Menezes Figueiredo, Rômulo Leão Silva Neris, Iris Paula Guimarães-Andrade, Daniel Gavino-Leopoldino, Leonardo Linhares Miler-da-Silva, Helber da Maia Valença, Leandro Ladislau, Caroline Victorino Felix de Lima, Fernanda Meireles Coccarelli, Claudia Farias Benjamim and Iranaia Assunção-Miranda
Int. J. Mol. Sci. 2024, 25(16), 8909; https://doi.org/10.3390/ijms25168909 - 16 Aug 2024
Viewed by 1279
Abstract
Chikungunya (CHIKV) and Mayaro (MAYV) viruses are arthritogenic alphaviruses that promote an incapacitating and long-lasting inflammatory muscle–articular disease. Despite studies pointing out the importance of skeletal muscle (SkM) in viral pathogenesis, the long-term consequences on its physiology and the mechanism of persistence of [...] Read more.
Chikungunya (CHIKV) and Mayaro (MAYV) viruses are arthritogenic alphaviruses that promote an incapacitating and long-lasting inflammatory muscle–articular disease. Despite studies pointing out the importance of skeletal muscle (SkM) in viral pathogenesis, the long-term consequences on its physiology and the mechanism of persistence of symptoms are still poorly understood. Combining molecular, morphological, nuclear magnetic resonance imaging, and histological analysis, we conduct a temporal investigation of CHIKV and MAYV replication in a wild-type mice model, focusing on the impact on SkM composition, structure, and repair in the acute and late phases of infection. We found that viral replication and induced inflammation promote a rapid loss of muscle mass and reduction in fiber cross-sectional area by upregulation of muscle-specific E3 ubiquitin ligases MuRF1 and Atrogin-1 expression, both key regulators of SkM fibers atrophy. Despite a reduction in inflammation and clearance of infectious viral particles, SkM atrophy persists until 30 days post-infection. The genomic CHIKV and MAYV RNAs were still detected in SkM in the late phase, along with the upregulation of chemokines and anti-inflammatory cytokine expression. In agreement with the involvement of inflammatory mediators on induced atrophy, the neutralization of TNF and a reduction in oxidative stress using monomethyl fumarate, an agonist of Nrf2, decreases atrogen expression and atrophic fibers while increasing weight gain in treated mice. These data indicate that arthritogenic alphavirus infection could chronically impact body SkM composition and also harm repair machinery, contributing to a better understanding of mechanisms of arthritogenic alphavirus pathogenesis and with a description of potentially new targets of therapeutic intervention. Full article
(This article belongs to the Special Issue Advanced Research on Immune Response to Viral Infection)
Show Figures

Figure 1

Figure 1
<p>Temporal investigation of CHIKV and MAYV replication and clearance from SkM after subcutaneous infection in a young wild-type mice model. Wild-type (WT) SV129 mice of 12 day-olds were subcutaneously infected with MAYV or CHIKV in the left footpad, and tissues were collected at indicated time points. (<b>A</b>) Temporal quantification of viral load by plaque assay in the left gastrocnemius, (<b>B</b>) distribution in other tissues with detected infectious particles at 4 dpi and (<b>C</b>) at 8 dpi. (<b>D</b>) Virus- and Mock-infected mice swelling area of left paws and (<b>E</b>) weight gain was accompanied temporally. (<b>F</b>) Gastrocnemius muscle was dissected and immediately weighed at 4 dpi. Values were plotted as mean ± standard error of the mean (SEM). The inset shows a representative image of dissected muscles. Statistical analyses were performed to compare (<b>A</b>–<b>C</b>) viral load of MAYV and CHIKV groups by multiple <span class="html-italic">t</span>-tests, and significance was determined using the Holm–Sidak method; (<b>D</b>,<b>E</b>) swelling and weight gain curve by two-way ANOVA and (<b>F</b>) muscle weight by one-way ANOVA followed by Tukey’s multiple comparison test from MAYV and CHIKV groups with mock. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001. Quad = quadriceps muscle. ND: not detected.</p>
Full article ">Figure 2
<p>MAYV and CHIKV replication induces inflammatory infiltration and lesions, muscle mass loss, and fiber atrophy in skeletal muscle. (<b>A</b>) Left gastrocnemius of Mock or infected groups were collected at 4 dpi and stained with hematoxylin and eosin (H&amp;E). Representative images from 2 infected mice demonstrate the inflammatory cell infiltration, fiber destruction, necrosis, and atrophic fibers in MAYV- and CHIKV-infected animals. High magnification images inset highlight the regions of atrophic fibers. Scale bars of figure = 100 μm and inset 50 μm. (<b>B</b>) Quantification of SkM fibers cross-sectional area (CSA) from H&amp;E stained muscle images was performed using ImageJ 1.52a software. Each dot corresponds to the average CSA of fibers present in a field per mouse. (<b>C</b>–<b>E</b>) The hind limbs of Mock- and MAYV-infected mice were imaged by NMRI. At each time point, images from five sections were acquired to calculate the area and then combined for volume reconstitution. (<b>F</b>) Expression levels of MuRF1 and Atrogin-1 in the left gastrocnemius at 4 dpi were determined by real-time PCR analysis. Cycle threshold (Ct) values were normalized to a housekeeping gene and analyzed using the ΔΔCt method to generate fold change values (2<sup>−ΔΔCt</sup>). Values are shown as mean ± standard error of the mean (SEM). Statistical analyses were performed using one-way ANOVA followed by Tukey’s multiple comparison tests (<b>B</b>,<b>F</b>), and for the comparison of tissue volumes of Mock and MAYV groups, multiple <span class="html-italic">t</span>-tests were used, and the significance was determined by the Holm–Sidak method. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001. Image (<b>C</b>) was created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 3
<p>Skeletal muscle atrophy and genomic RNA persist in the late phase of MAYV and CHIKV infection. Wild-type (WT) SV129 mice of 12-day-olds were infected, and SkMs were analyzed at late times post-infection. (<b>A</b>) Body weight was recorded until 30 dpi, and (<b>B</b>) gastrocnemius muscles were dissected and immediately weighed. (<b>C</b>) CHIKV and MAYV RNA were detected by TaqMan real-time PCR analysis. The dotted lines represent the Ct value limit for positive samples. (<b>D</b>) Temporal histological analyses of left gastrocnemius stained with H&amp;E and respective quantification of SkM fiber cross-sectional area (CSA) from H&amp;E stained muscle images using ImageJ 1.52a software. Each dot corresponds to the average CSA of fibers present in a field per mouse. Scale bars of figure = 100 μm and inset 50 μm. Values are shown as mean ± standard error of the mean (SEM). Statistical analysis of the weight gain curve comparing MAYV and CHIKV groups with Mock was performed by two-way ANOVA (<b>A</b>); for muscle weight (<b>B</b>) and CSA (<b>D</b>), one-way ANOVA was performed, followed by Tukey’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 for Mock and MAYV comparison; <span class="html-italic"><sup>#</sup> p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 for Mock and CHIKV comparison.</p>
Full article ">Figure 4
<p>Temporal analysis of inflammatory mediator and atrogen expression in the early and late phases of MAYV and CHIKV infection. Wild-type (WT) SV129 mice of 12 day-olds were subcutaneously infected with MAYV or CHIKV in the left footpad, and left gastrocnemius were collected at indicated time points. (<b>A</b>–<b>I</b>,<b>K</b>,<b>L</b>) Quantification of gene expression using real-time PCR analysis. Cycle threshold (Ct) values were normalized to a housekeeping gene and analyzed using the ΔΔCt method to generate fold change values (2<sup>−ΔΔCt</sup>). (<b>J</b>) Total reactive oxygen species (ROS) production in the left gastrocnemius was determined by fluorescence analysis using DCFDA. Arbitrary values of fluorescence from each sample were obtained at the end of the DCF incubation and plotted as fold change from the media of mock values. Values were plotted as mean ± standard error of the mean (SEM). Statistical analyses were performed by multiple <span class="html-italic">t</span>-tests to compare Mock with CHIKV and MAYV groups, and the significance was determined using the Holm–Sidak method. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Treatment with Inflixmab reduces muscle mass loss and atrogen expression induced by CHIKV infection. Wild-type (WT) SV129 mice of 12 day-olds were subcutaneously infected with CHIKV and then treated with a daily dose of 20 μg of Infliximab (IFX) or Vehicle (PBS) by i.p. inoculation. (<b>A</b>) Weight gain was accompanied daily, and (<b>B</b>) gastrocnemius was dissected at 4 dpi for muscle weight measurement, (<b>C</b>) Atrogin expression by real-time PCR, and (<b>D</b>) viral load by plaque assay. (<b>E</b>) Virus- and Mock-infected mice swelling area of left paws was measured daily. Values were plotted as mean ± standard error of the mean (SEM). Statistical analyses of weight gain curve and paw swelling comparing treated and untreated CHIKV infected groups were performed by two-way ANOVA (<b>A</b>,<b>E</b>); and for muscle weight (<b>B</b>), Atrogin-1 expression (<b>C</b>), and viral load (<b>D</b>), by one-way ANOVA followed by Tukey’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Treatment with MMF reduces muscle atrophy and inflammation induced by MAYV and CHIKV infection. Wild-type (WT) SV129 mice of 12 day-olds were intraperitoneally treated with 20 mg/kg of monomethyl fumarate (MMF) or Vehicle (DMSO) and after 4 h infected with MAYV or CHIKV in the left footpad. (<b>A</b>) Weight gain was accompanied daily. Gastrocnemius were dissected at 8 dpi for analysis of (<b>B</b>) muscle weight, (<b>C</b>) MuRF1, and (<b>D</b>) Atrogin-1 expression by real-time PCR. (<b>E</b>) Histological analysis of left gastrocnemius stained with H&amp;E (Scale bars of figures = 100 μm) and (<b>F</b>) respective quantification of SkM fiber cross-sectional area (CSA) from muscle images using ImageJ 1.52a software. (<b>G</b>) Viral load at SkM was determined by plaque assay at 4 and 8 dpi. Values were plotted as mean ± standard error of the mean (SEM). ND: not detected. Statistical analyses of weight gain curve comparing treated and untreated CHIKV and MAYV infected groups were performed by two-way ANOVA (<b>A</b>) with * used for indicates significance of comparison of both MAYV and CHIKV groups (4 and 6 dpi); # Only MAYV, and &amp; CHIKV groups (8 dpi); one-way ANOVA was used in the analysis of muscle weight (<b>B</b>), Atrogens expression (<b>C,D</b>), CSA (<b>F</b>), and viral load (<b>G</b>), followed by Tukey’s multiple comparison test. */<sup>&amp;</sup> <span class="html-italic">p</span> &lt; 0.05, **/<sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Schematic representation of the possible mechanism involved in arthritogenic alphavirus-induced early and late skeletal muscle atrophy. ① At physiological conditions, young mice SkM fiber growth throughout life is determined by individual muscle fiber enlargement and mass gain by a higher rate of synthesis than protein degradation. ② New fiber generation by myogenesis will be recruited mainly after injury by activation of quiescent satellite cells. ③–⑧ Arthritogenic alphavirus replicates in SkM fibers, resulting in fiber destruction, recruitment of immune cells, and the production of inflammatory mediators in the early phase of infection. ⑨–⑩ Inflammation-induced protein degradation by UPS through activation of MuRF1 and Atrogin-1 that drives acute SkM atrophy. ⑪–⑫ Treatment with IFX or MMF reduces protein degradation and atrophy, corroborating the involvement of these pathways in MAYV- and CHIKV-induced atrophy. ⑬ SkM atrophy persists in the late phase of infection. ⑭ Despite the decrease in TNF, IFNγ, and IL-6 and also atrogen expression levels, ⑮–⑰ chemokines, IL-10, TGFβ mediators, and viral genomic RNA are still detected, indicating an incomplete viral clearance. ⑱ Long-term immune activation results in a reduction in CSA by a UPS-independent mechanism. Symbols indicates: <span class="html-fig-inline" id="ijms-25-08909-i001"><img alt="Ijms 25 08909 i001" src="/ijms/ijms-25-08909/article_deploy/html/images/ijms-25-08909-i001.png"/></span> block/inhibition; <span class="html-fig-inline" id="ijms-25-08909-i002"><img alt="Ijms 25 08909 i002" src="/ijms/ijms-25-08909/article_deploy/html/images/ijms-25-08909-i002.png"/></span> positive regulation; <span class="html-fig-inline" id="ijms-25-08909-i003"><img alt="Ijms 25 08909 i003" src="/ijms/ijms-25-08909/article_deploy/html/images/ijms-25-08909-i003.png"/></span> open ended questions.</p>
Full article ">
24 pages, 1798 KiB  
Review
New Trends to Treat Muscular Atrophy: A Systematic Review of Epicatechin
by Iris Jasmin Santos German, Karina Torres Pomini, Jesus Carlos Andreo, João Vitor Tadashi Cosin Shindo, Marcela Vialogo Marques de Castro, Claudia Rucco P. Detregiachi, Adriano Cressoni Araújo, Elen Landgraf Guiguer, Lucas Fornari Laurindo, Patrícia Cincotto dos Santos Bueno, Maricelma da Silva Soares de Souza, Marcia Gabaldi, Sandra Maria Barbalho and André Luis Shinohara
Nutrients 2024, 16(2), 326; https://doi.org/10.3390/nu16020326 - 22 Jan 2024
Cited by 5 | Viewed by 6585
Abstract
Epicatechin is a polyphenol compound that promotes skeletal muscle differentiation and counteracts the pathways that participate in the degradation of proteins. Several studies present contradictory results of treatment protocols and therapeutic effects. Therefore, the objective of this systematic review was to investigate the [...] Read more.
Epicatechin is a polyphenol compound that promotes skeletal muscle differentiation and counteracts the pathways that participate in the degradation of proteins. Several studies present contradictory results of treatment protocols and therapeutic effects. Therefore, the objective of this systematic review was to investigate the current literature showing the molecular mechanism and clinical protocol of epicatechin in muscle atrophy in humans, animals, and myoblast cell-line. The search was conducted in Embase, PubMed/MEDLINE, Cochrane Library, and Web of Science. The qualitative analysis demonstrated that there is a commonness of epicatechin inhibitory action in myostatin expression and atrogenes MAFbx, FOXO, and MuRF1. Epicatechin showed positive effects on follistatin and on the stimulation of factors related to the myogenic actions (MyoD, Myf5, and myogenin). Furthermore, the literature also showed that epicatechin can interfere with mitochondrias’ biosynthesis in muscle fibers, stimulation of the signaling pathways of AKT/mTOR protein production, and amelioration of skeletal musculature performance, particularly when combined with physical exercise. Epicatechin can, for these reasons, exhibit clinical applicability due to the beneficial results under conditions that negatively affect the skeletal musculature. However, there is no protocol standardization or enough clinical evidence to draw more specific conclusions on its therapeutic implementation. Full article
(This article belongs to the Special Issue Nutritional Strategies for Diseases Associated with Muscle Injuries)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Main food and beverage sources, chemical structure, metabolic route, and biological properties of epicatechin. NO, nitric oxide; Nrf2, nuclear factor-like 1; HO, heme oxygenase; PGC1α, peroxisome proliferator-activated receptor γ coactivator 1-alpha; ROS, reactive oxygen species.</p>
Full article ">Figure 2
<p>PRISMA 2020 flow diagram. Search design strategy in the databases and other sources. **: records identified in the databases but not related to this review topic.</p>
Full article ">Figure 3
<p>An overview of epicatechin effects on skeletal muscle. Modified diagram from Li et al. [<a href="#B4-nutrients-16-00326" class="html-bibr">4</a>]. Act RIIB, myostatin receptor; Smad 2/3, mothers against decapentaplegic homolog 2; FoxO, forkhead transcription factor family; MAFbx, muscle atrophy F-box; MuRF1, muscle RING-finger protein-1; IGF, insulin-like growth factor-1; PI3K, phosphatidylinositol 3 kinase 9; AKT, Protein kinase B; mTOR, The mammalian target of rapamycin.</p>
Full article ">
16 pages, 3012 KiB  
Article
Effects of Tofacitinib on Muscle Remodeling in Experimental Rheumatoid Sarcopenia
by Ismael Bermejo-Álvarez, Sandra Pérez-Baos, Paula Gratal, Juan Pablo Medina, Raquel Largo, Gabriel Herrero-Beaumont and Aránzazu Mediero
Int. J. Mol. Sci. 2023, 24(17), 13181; https://doi.org/10.3390/ijms241713181 - 24 Aug 2023
Cited by 5 | Viewed by 2348
Abstract
Sarcopenia is a frequent comorbidity of rheumatoid arthritis (RA). Clinical trials have shown that JAK inhibitors (JAKi) produce an asymptomatic increase in serum creatine kinase (CK) in RA, suggesting an impact on muscle. We evaluated the effect of JAKi in muscle remodeling in [...] Read more.
Sarcopenia is a frequent comorbidity of rheumatoid arthritis (RA). Clinical trials have shown that JAK inhibitors (JAKi) produce an asymptomatic increase in serum creatine kinase (CK) in RA, suggesting an impact on muscle. We evaluated the effect of JAKi in muscle remodeling in an experimental RA model. Antigen-induced arthritis (experimental RA, e-RA) was performed in 14 rabbits. Seven rabbits received tofacitinib (TOFA, orally 10 mg/kg/day). Animals were euthanized one day after the last ovalbumin injection, and muscles were prepared for histology, RT-PCR, and WB. C-reactive protein (CRP) and Myostatin (MSTN) serum concentration were determined by ELISA. Creatine and creatine kinase (CK) were analyzed. An increase in body weight as well as tibialis anterior cross-sectional area and diameter was observed in e-RA+TOFA vs. e-RA. e-RA decreased type II fibers and increased the myonuclei number, with all reverted by TOFA. TOFA did not modify CRP levels, neither did MSTN. TOFA significantly reduced IL-6, atrogin-1, and MuRF-1 compared with e-RA. e-RA+TOFA showed higher CK and lower creatine levels compared with e-RA. No differences in PAX-7 were found, while TOFA prevented the increase in MyoD1 in e-RA. Our model reflects the features of rheumatoid sarcopenia in RA. JAKi increased muscle mass through attenuating IL-6/JAK/STAT activation, decreasing atrogenes, and restoring muscle differentiation markers. These data together with an increase in CK support the role of CK as a valuable marker of muscle gain following JAKi treatment. Full article
(This article belongs to the Special Issue Molecular and Therapeutic Research in Rheumatoid Arthritis)
Show Figures

Figure 1

Figure 1
<p>Experimental model of rheumatoid arthritis in rabbits that replicate human disease. (<b>A</b>) Chronogram of the experimental model. (<b>B</b>) Body weight gain (kg) of rabbits at the end of the study. (<b>C</b>) Serum CRP levels (μg/mL) at the end of the study. Data are shown as the mean and SEM (<span class="html-italic">n</span> = 7 rabbits per group). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. healthy # <span class="html-italic">p</span> &lt; 0.05 vs. CRP: C-reactive protein; e-RA: experimental rheumatoid arthritis; OVA: ovalbumin; TOFA: tofacitinib.</p>
Full article ">Figure 2
<p>TOFA improves structural skeletal muscle changes. (<b>A</b>) Tibialis anterior (TA) weight (g). (<b>B</b>) Representative TA from healthy control and e-RA groups. Posterior view, scale bar = 1 cm. (<b>C</b>) Cross-sectional diameter of TA (mm) and cross-sectional area of TA (mm<sup>2</sup>). (<b>D</b>) Representative mid-belly cross sections of TA in control and e-RA groups stained with H&amp;E. Scale bar = 2.5 mm. (<b>E</b>) Representative focal microscopy images from segments of extensor digitorium longus (EDL) fibers in healthy and e-RA groups. Rhodamine phalloidin (red) was used to stain actin and DAPI (blue) to stain nuclei. Scale bar = 30 μm. Number of myonuclei in healthy in comparison with e-RA groups expressed as number of nuclei per 100 mm segment. (<b>F</b>) Representative sections of type I (white) and II (black) fiber distribution and size in TA of healthy and e-RA groups stained with ATPase pH 9.4. Scale bar = 100 μm. Data are shown as the mean and SEM (<span class="html-italic">n</span> = 7 rabbits per group). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. healthy; # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, and ### <span class="html-italic">p</span> &lt; 0.001 vs. e-RA. EDL: extensor digitorum longus; e-RA: experimental rheumatoid arthritis; TA: tibialis anterior; TOFA: tofacitinib.</p>
Full article ">Figure 3
<p>No differences in PAX-7 and MSTN were found, while TOFA tended to revert MyoD1 expression. (<b>A</b>) Densitometric analysis of Pax7, MyoD1, and Myogenin protein expression levels in gastrocnemius. Data are normalized to endogenous control (α-tubulin) and expressed as arbitrary units (A.U.). Representative cropped blots of two animals of each group are shown: healthy, e-RA, and e-RA+TOFA, respectively. (<b>B</b>) Gene and serum expression of MSTN. Data are shown as the mean and SEM (<span class="html-italic">n</span> = 7 rabbits per group). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. healthy. e-RA: experimental rheumatoid arthritis; MSTN: Myostatin; TOFA: tofacitinib.</p>
Full article ">Figure 4
<p>Pro-inflammatory mediators and atrogenes. (<b>A</b>) Gene expression for IL-1β, IL-6, and TNFα. (<b>B</b>) Gene expression for atrogin-1 and MuRF1. (<b>C</b>) Gene expression for MCP-1. Data are shown as the mean and SEM (<span class="html-italic">n</span> = 7 rabbits per group). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. healthy; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. e-RA. e-RA: experimental rheumatoid arthritis; IL-1: interleukin 1β; MCP-1: monocyte chemotactic protein-1; MuRF1: muscle RING-finger protein-1; TOFA: tofacitinib; TNF: tumor necrosis factor.</p>
Full article ">Figure 5
<p>TOFA showed increased creatine kinase and lower creatine in muscle. (<b>A</b>) Serum and tissue CK levels. (<b>B</b>) Creatine (ng/mg tissue) and pyruvate (nmol/mg protein) in muscle. Data are shown as the mean and SEM (<span class="html-italic">n</span> = 7 rabbits per group). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 vs. healthy; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 vs. e-RA. CK: creatine kinase; e-RA: experimental rheumatoid arthritis; TOFA: tofacitinib.</p>
Full article ">Figure 6
<p>TOFA decreases STAT3 but not STAT1 signaling. (<b>A</b>) Densitometric analysis of pSTAT1 and pSTAT3 protein expression in gastrocnemius. (<b>B</b>) Densitometric analysis of SOCS1 and SOCS3 protein expression in gastrocnemius. Data are normalized to endogenous control (α-tubulin) and expressed as arbitrary units (A.U.). Representative cropped blots of two animals of each group are shown: healthy, e-RA, and e-RA+TOFA, respectively. Data are shown as the mean and SEM (<span class="html-italic">n</span> = 7 rabbits per group). *** <span class="html-italic">p</span> &lt; 0.001 vs. healthy. e-RA: experimental rheumatoid arthritis; TOFA: tofacitinib.</p>
Full article ">
9 pages, 1417 KiB  
Editorial
Introduction to the Special Issue “Skeletal Muscle Atrophy: Mechanisms at a Cellular Level”
by Emanuela Zuccaro, Caterina Marchioretti, Marco Pirazzini and Maria Pennuto
Cells 2023, 12(3), 502; https://doi.org/10.3390/cells12030502 - 3 Feb 2023
Cited by 1 | Viewed by 5054
Abstract
Skeletal muscle is the most abundant tissue in the body and requires high levels of energy to function properly. Skeletal muscle allows voluntary movement and body posture, which require different types of fiber, innervation, energy, and metabolism. Here, we summarize the contribution received [...] Read more.
Skeletal muscle is the most abundant tissue in the body and requires high levels of energy to function properly. Skeletal muscle allows voluntary movement and body posture, which require different types of fiber, innervation, energy, and metabolism. Here, we summarize the contribution received at the time of publication of this Introductory Issue for the Special Issue dedicated to “Skeletal Muscle Atrophy: Mechanisms at a Cellular Level”. The Special Issue is divided into three sections. The first is dedicated to skeletal muscle pathophysiology, the second to disease mechanisms, and the third to therapeutic development. Full article
(This article belongs to the Special Issue Skeletal Muscle Atrophy: Mechanisms at a Cellular Level)
Show Figures

Figure 1

Figure 1
<p>Structure of skeletal muscle, myofibers, and the sarcomere. The muscle is made up of several myofibrils packed into functional units surrounded by different layers of connective tissues (epimysium, perimysium and endomysium). One myofiber is composed of several sarcomeres, which is the main contractile unit mainly composed of protein filaments (myofilaments), namely myosin (thick filaments) and actin (thin filaments). The structure of the sarcomere presents dark and light bands, also visible with a light microscope. This is due to the alternated arrangement of the A bands (or anisotropic bands), the dark bands containing whole thick filaments, and the I bands (or isotropic bands), the light bands made up of only the thin filaments, located between two thick filaments. The Z disc across I bands demarcates the point of attachment between two adjacent actin filaments. The M line is considered the center of a sarcomere, whereas the H zone, which contains only myosin, is the area in between the M line and Z disc in resting conditions. Each myofibril is associated with troponin and tropomyosin, two regulatory proteins of muscle contraction.</p>
Full article ">Figure 2
<p>From neurotransmitter release to muscle contraction. The action potential running down to the motor axon invades the nerve terminal at the neuromuscular junction and triggers the fusion of the synaptic vesicles with the presynaptic membrane. Acetylcholine (ACh) is thus released and diffuses in the synaptic cleft to bind to the nicotinic ACh receptors expressed on the postsynaptic muscle fiber membrane. nAChRs are ionotropic ligand-gated Na<sup>+</sup>/K<sup>+</sup> channels that cause a local depolarization of the muscle end-plate via inward flux of Na<sup>+</sup>. When the depolarization overcomes a critical threshold, voltage-gated Na<sup>+</sup> channels open, thus triggering a post-synaptic action potential into the muscle fiber, which ultimately spreads out along the sarcolemma and invades the T-tubules (brown arrows). Here, an excitation-contraction molecular machinery transduces this electric signal into the cytosolic release of Ca<sup>2+</sup> from the sarcoplasmic reticulum, leading to muscle fiber contraction.</p>
Full article ">
18 pages, 3100 KiB  
Article
Induction of ATF4-Regulated Atrogenes Is Uncoupled from Muscle Atrophy during Disuse in Halofuginone-Treated Mice and in Hibernating Brown Bears
by Laura Cussonneau, Cécile Coudy-Gandilhon, Christiane Deval, Ghita Chaouki, Mehdi Djelloul-Mazouz, Yoann Delorme, Julien Hermet, Guillemette Gauquelin-Koch, Cécile Polge, Daniel Taillandier, Julien Averous, Alain Bruhat, Céline Jousse, Isabelle Papet, Fabrice Bertile, Etienne Lefai, Pierre Fafournoux, Anne-Catherine Maurin and Lydie Combaret
Int. J. Mol. Sci. 2023, 24(1), 621; https://doi.org/10.3390/ijms24010621 - 30 Dec 2022
Cited by 2 | Viewed by 2380
Abstract
Activating transcription factor 4 (ATF4) is involved in muscle atrophy through the overexpression of some atrogenes. However, it also controls the transcription of genes involved in muscle homeostasis maintenance. Here, we explored the effect of ATF4 activation by the pharmacological molecule halofuginone during [...] Read more.
Activating transcription factor 4 (ATF4) is involved in muscle atrophy through the overexpression of some atrogenes. However, it also controls the transcription of genes involved in muscle homeostasis maintenance. Here, we explored the effect of ATF4 activation by the pharmacological molecule halofuginone during hindlimb suspension (HS)-induced muscle atrophy. Firstly, we reported that periodic activation of ATF4-regulated atrogenes (Gadd45a, Cdkn1a, and Eif4ebp1) by halofuginone was not associated with muscle atrophy in healthy mice. Secondly, halofuginone-treated mice even showed reduced atrophy during HS, although the induction of the ATF4 pathway was identical to that in untreated HS mice. We further showed that halofuginone inhibited transforming growth factor-β (TGF-β) signalling, while promoting bone morphogenetic protein (BMP) signalling in healthy mice and slightly preserved protein synthesis during HS. Finally, ATF4-regulated atrogenes were also induced in the atrophy-resistant muscles of hibernating brown bears, in which we previously also reported concurrent TGF-β inhibition and BMP activation. Overall, we show that ATF4-induced atrogenes can be uncoupled from muscle atrophy. In addition, our data also indicate that halofuginone can control the TGF-β/BMP balance towards muscle mass maintenance. Whether halofuginone-induced BMP signalling can counteract the effect of ATF4-induced atrogenes needs to be further investigated and may open a new avenue to fight muscle atrophy. Finally, our study opens the way for further studies to identify well-tolerated chemical compounds in humans that are able to fine-tune the TGF-β/BMP balance and could be used to preserve muscle mass during catabolic situations. Full article
(This article belongs to the Special Issue Muscle Atrophy: From Bench to Bedside)
Show Figures

Figure 1

Figure 1
<p>Halofuginone activates the expression of ATF4-regulated atrogenes in muscle without leading to atrophy. (<b>A</b>) Schematic representation of the experimental protocol, where mice received H<sub>2</sub>O (white bars) or HF (0.25 µg/g, grey bars) 3 times a week for up to 4 weeks (WK). Muscles were collected 6 h after the last HF administration at the end of each week (dotted arrows). (<b>B</b>–<b>F</b>) Relative mRNA levels in gastrocnemius for <span class="html-italic">Atf4</span>, <span class="html-italic">Trib3</span>, <span class="html-italic">Cdkn1a</span>, <span class="html-italic">Gadd45a</span>, and <span class="html-italic">Eif4ebp1</span> were measured by RT-qPCR. Data were normalised using <span class="html-italic">Tbp</span>. Data are expressed as fold change vs. H<sub>2</sub>O within each week and are presented as individual values with mean bars ± SEM. (<b>G</b>) Gastrocnemius muscle mass per gram of body weight (BW). Data are expressed as a percentage from H<sub>2</sub>O within each week and presented as individual values with mean bars ± SEM. Statistics are described in <a href="#sec4-ijms-24-00621" class="html-sec">Section 4</a>. * <span class="html-italic">p</span><sub>adj</sub> &lt; 0.05; ** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.01; *** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.001; **** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Hindlimb suspension induces ATF4 pathway. (<b>A</b>) Schematic representation of the experimental protocol, where mice received H<sub>2</sub>O or halofuginone (HF) oral administration (0.25 µg/g) 3 times a week for 3 weeks (WK) (black arrows) and were then subjected to hindlimb suspension for 3 (HS3, light grey bars) or 7 (HS7, white bars) days or kept unsuspended (Ctrl, dark grey). The dotted arrows represent the time when the muscles were collected. (<b>B</b>–<b>D</b>) Relative protein levels in gastrocnemius for phosphorylated and total eIF2α were measured by Western blotting, quantified, and normalised to the total protein content. Representative Western blots are shown. (<b>E</b>–<b>I</b>) Relative mRNA levels in gastrocnemius for <span class="html-italic">Atf4</span>, <span class="html-italic">Trib3</span>, <span class="html-italic">Cdkn1a, Eif4ebp1</span>, and <span class="html-italic">Gadd45a</span> were measured by RT-qPCR and were normalised using <span class="html-italic">Tbp.</span> Data are expressed as fold change vs. H<sub>2</sub>O-Ctrl and are presented as individual values normalised mean bars ± SEM. Statistics are described in <a href="#sec4-ijms-24-00621" class="html-sec">Section 4</a>. * <span class="html-italic">p</span><sub>adj</sub> &lt; 0.05; ** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.01; *** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.001; **** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Halofuginone treatment prior to hindlimb suspension mitigates atrophy in gastrocnemius muscle. Mice were treated with H<sub>2</sub>O or halofuginone (HF, 0.25 µg/g) 3 times a week for 3 weeks and were then subjected to hindlimb suspension for 3 (HS3, light grey bars) or 7 (HS7, white bars) days or kept unsuspended (Ctrl, dark grey bars), as described in <a href="#ijms-24-00621-f002" class="html-fig">Figure 2</a>A. (<b>A</b>) Gastrocnemius muscle mass per gram of body weight (BW). Data are expressed as a percentage from H<sub>2</sub>O-Ctrl and presented as individual values with mean bars ± SEM. (<b>B</b>) Mean fibre cross-sectional area in gastrocnemius muscle. Data are presented as individual values with mean bars ± SEM. Statistics are described in <a href="#sec4-ijms-24-00621" class="html-sec">Section 4</a>. ** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.01; *** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.001; **** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.0001; ns = non-significant.</p>
Full article ">Figure 4
<p>Halofuginone treatment inhibits TGF-β while promoting BMP signalling in gastrocnemius muscle. Mice were treated with H<sub>2</sub>O or halofuginone (HF, 0.25 µg/g) 3 times a week for 3 weeks and were then subjected to hindlimb suspension for 3 (HS3, light grey bars) or 7 (HS7, white bars) days or kept unsuspended (Ctrl, dark grey bars), as described in <a href="#ijms-24-00621-f002" class="html-fig">Figure 2</a>A. (<b>A</b>–<b>D</b>) The ratio of protein levels in gastrocnemius for the transcription factors SMAD2/3 (TGF-β signalling), SMAD1/5 (BMP signalling), and SMAD4 (TGF-β and BMP signalling) have been assessed in the nuclear and cytosolic subcellular fractions, quantified, and normalised to the total protein content. Representative Western blots are shown. The ratio of nuclear SMAD contents on the total (cytosolic and nuclear) SMAD content was calculated. Data are expressed as fold change vs. H<sub>2</sub>O-Ctrl and presented as individual values with mean bars ± SEM. Statistics are described in <a href="#sec4-ijms-24-00621" class="html-sec">Section 4</a>. * <span class="html-italic">p</span><sub>adj</sub> &lt; 0.05; **** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Halofuginone treatment prior to hindlimb suspension partially prevents the decrease in protein synthesis in gastrocnemius muscle. Mice were treated with H<sub>2</sub>O or halofuginone (HF, 0.25 µg/g) 3 times a week for 3 weeks and were then subjected to hindlimb suspension for 3 (HS3, light grey bars) or 7 (HS7, white bars) days or kept unsuspended (Ctrl, dark grey bars), as described in <a href="#ijms-24-00621-f002" class="html-fig">Figure 2</a>A. (<b>A</b>,<b>B</b>) Relative puromycin incorporation into gastrocnemius muscle was assessed by Western blotting, quantified, and normalised to the total protein content. A representative Western blot is shown. (<b>C</b>–<b>E</b>) Relative mRNA levels in gastrocnemius for <span class="html-italic">Trim63</span>, <span class="html-italic">Fbxo32</span>, and <span class="html-italic">Fbxo30</span> were measured by RT-qPCR. Data were normalised using <span class="html-italic">Tbp</span>. Data are expressed as fold change vs. H<sub>2</sub>O-Ctrl and presented as individual values with mean bars ± SEM. Statistics are described in <a href="#sec4-ijms-24-00621" class="html-sec">Section 4</a>. * <span class="html-italic">p</span><sub>adj</sub> &lt; 0.05; **** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.0001, or ns = non-significant.</p>
Full article ">Figure 6
<p>ATF4-regulated atrogenes are induced in atrophy-resistant hibernating brown bear muscles. Gene expression levels for <span class="html-italic">ATF4</span>, <span class="html-italic">GADD45A</span>, <span class="html-italic">CDKN1A</span>, <span class="html-italic">TRIB3</span>, <span class="html-italic">EIF4EBP1</span>, <span class="html-italic">PPP1R15A</span>, <span class="html-italic">ASNS</span>, and <span class="html-italic">DDIT3</span> in vastus lateralis muscle of active and hibernating brown bears (n = 6 bears/season, the same individuals were sampled and analysed in summer and winter, log2FC winter/summer). Data are presented as individual values as log2FC with mean bars ± lfcSE (log2 fold change standard error). Statistics are described in [<a href="#B46-ijms-24-00621" class="html-bibr">46</a>]. * <span class="html-italic">p</span><sub>adj</sub> &lt; 0.05; ** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.01; **** <span class="html-italic">p</span><sub>adj</sub> &lt; 0.0001. FC: fold change; W: winter (hibernating season); S: summer (active season).</p>
Full article ">Figure 7
<p>Graphical abstract. The red and green lines represent catabolic and anabolic effects, respectively. Dotted lines represent hypothetical connections. The arrows/T bars above the ATF4 atrogenes, SMAD2/3, and SMAD1/5 boxes represent the induction/inhibition by halofuginone or by an as-yet-unknown mechanism in mouse or bear muscle, respectively. Created with BioRender.com.</p>
Full article ">
12 pages, 674 KiB  
Review
Mammalian Target of Rapamycin (mTOR) Signaling at the Crossroad of Muscle Fiber Fate in Sarcopenia
by Giuseppe Sirago, Anna Picca, Riccardo Calvani, Hélio José Coelho-Júnior and Emanuele Marzetti
Int. J. Mol. Sci. 2022, 23(22), 13823; https://doi.org/10.3390/ijms232213823 - 10 Nov 2022
Cited by 21 | Viewed by 5492
Abstract
The mammalian target of rapamycin (mTOR) is a major regulator of skeletal myocyte viability. The signaling pathways triggered by mTOR vary according to the type of endogenous and exogenous factors (e.g., redox balance, nutrient availability, physical activity) as well as organismal age. Here, [...] Read more.
The mammalian target of rapamycin (mTOR) is a major regulator of skeletal myocyte viability. The signaling pathways triggered by mTOR vary according to the type of endogenous and exogenous factors (e.g., redox balance, nutrient availability, physical activity) as well as organismal age. Here, we provide an overview of mTOR signaling in skeletal muscle, with a special focus on the role played by mTOR in the development of sarcopenia. Intervention strategies targeting mTOR in sarcopenia (e.g., supplementation of plant extracts, hormones, inorganic ions, calorie restriction, and exercise) have also been discussed. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Schematic Representation of the Coordinated Regulation of the Mammalian Target of Rapamycin and Mitophagy in Muscle Protein Synthesis and Degradation. In the presence of growth factors (e.g., insulin, insulin-like growth factor 1, growth hormone), the phosphoinositide 3-kinases-protein kinase B (PI3K) is activated and triggers muscle protein synthesis via the mammalian target of rapamycin complex 1 (mTORC1). The latter complex is also positively modulated by amino acid availability. Conversely, downregulation of PI3K signaling induces translocation of Forkhead box O (FoxO) into the nucleus, where it regulates the transcription of the ubiquitin-ligases muscle ring finger 1 (MuRF1) and muscle atrophy F-box (MAFbx) genes. The activation of this signaling pathway induces degradation of sarcomere components and ignites a muscle pro-atrophy response. The same degradative molecular program is also triggered by fibroblast growth factor 21 (FGF21) in the setting of mitochondrial dysfunction and oxidative stress. In this case, the release of FGF21 stimulates the expression of the mitophagy-related protein B-cell lymphoma 2 interacting protein 3, paralleled by recruitment of the phosphatase and tensin homolog-induced kinase 1 (PINK1) through the translocases of the inner and the outer membranes and its activation at the site of depolarized mitochondria. This event promotes the sequestration of the E3 ubiquitin ligase Parkin at the outer mitochondrial membrane and guides the clearance of dysfunctional organelles. Finally, depolarized mitochondria are coated and prepared for disposal by the ubiquitin-binding adaptor protein p62/sequestosome-1 and the recruitment of the microtubule-associated proteins 1A/1B light chain 3B (LC3). This enables the transfer of mitochondria to lysosomes. FoxO-dependent atrophy is also pursued when severely damaged and bioenergetically incompetent mitochondria are not efficiently removed and, thus, the AMP/ATP ratio increases, which engages 5′ AMP-activated protein kinase (AMPK). Abbreviations: AA, amino acid; AMP, adenosine monophosphate; ATP, adenosine triphosphate; GF, growth factor; TIM23, translocase of the inner membrane 23; TOM, translocase of the outer membrane. Created with BioRender.com, accessed on 26 August 2022.</p>
Full article ">
20 pages, 7304 KiB  
Article
Coapplication of Magnesium Supplementation and Vibration Modulate Macrophage Polarization to Attenuate Sarcopenic Muscle Atrophy through PI3K/Akt/mTOR Signaling Pathway
by Can Cui, Zhengyuan Bao, Simon Kwoon-Ho Chow, Ronald Man Yeung Wong, Ailsa Welch, Ling Qin and Wing Hoi Cheung
Int. J. Mol. Sci. 2022, 23(21), 12944; https://doi.org/10.3390/ijms232112944 - 26 Oct 2022
Cited by 14 | Viewed by 3476
Abstract
Sarcopenia is an age-related geriatric syndrome characterized by the gradual loss of muscle mass and function. Low-magnitude high-frequency vibration (LMHFV) was shown to be beneficial to structural and functional outcomes of skeletal muscles, while magnesium (Mg) is a cofactor associated with better indices [...] Read more.
Sarcopenia is an age-related geriatric syndrome characterized by the gradual loss of muscle mass and function. Low-magnitude high-frequency vibration (LMHFV) was shown to be beneficial to structural and functional outcomes of skeletal muscles, while magnesium (Mg) is a cofactor associated with better indices of skeletal muscle mass and strength. We hypothesized that LMHFV, Mg and their combinations could suppress inflammation and sarcopenic atrophy, promote myogenesis via PI3k/Akt/mTOR pathway in senescence-accelerated mouse P8 (SAMP8) mice and C2C12 myoblasts. Results showed that Mg treatment and LMHFV could significantly decrease inflammatory expression (C/EBPα and LYVE1) and modulate a CD206-positive M2 macrophage population at month four. Mg treatment also showed significant inhibitory effects on FOXO3, MuRF1 and MAFbx mRNA expression. Coapplication showed a synergistic effect on suppression of type I fiber atrophy, with significantly higher IGF-1, MyoD, MyoG mRNA (p < 0.05) and pAkt protein expression (p < 0.0001) during sarcopenia. In vitro inhibition of PI3K/Akt and mTOR abolished the enhancement effects on myotube formation and inhibited MRF mRNA and p85, Akt, pAkt and mTOR protein expressions. The present study demonstrated that the PI3K/Akt/mTOR pathway is the predominant regulatory mechanism through which LMHFV and Mg enhanced muscle regeneration and suppressed atrogene upregulation. Full article
(This article belongs to the Special Issue Skeletal Muscle and Metabolic Disorders)
Show Figures

Figure 1

Figure 1
<p>Body mass parameters by Dual Energy X-rat Absorptiometry (DXA) and serum Mg concentration of SAMP8 mice. (<b>A</b>) Serum Mg levels in Con group showed a decreasing trend from month 2 to month 4, and serum Mg concentration in the Mg group was significantly higher than in the Con group at month 4. (<b>B</b>) Representative DXA images in lean mass mode. (<b>C</b>) Lean mass, (<b>D</b>) lean mass percentage, (<b>E</b>) appendicular lean mass and (<b>F</b>) appendicular lean mass percentage of SAMP8 mice in different groups at month 2, 3 and 4 post treatment. Lean mass in the Mg group was significantly higher than in the Con group at month 4. Mg + VIB group showed a significantly higher lean mass/percentage and appendicular lean mass/percentage compared with the Con group at month 2, and the VIB group showed significantly higher mass/percentage and appendicular lean mass/percentage compared with the Con group at month 3 and significantly higher lean mass percentage and percentage lean mass at month 4. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, n = 3–5.</p>
Full article ">Figure 2
<p>Immunohistochemical staining of myofibers (myosin heavy chain expression) in different groups at month 2, 3 and 4 post treatment. (<b>A</b>) Type I muscle fibers (blue), type IIa muscle fibers (green) and type IIb muscle fibers (red) of gastrocnemius at month 2, 3 and 4. (<b>B</b>–<b>D</b>) Type I, IIa and IIb CSA and percentage area in different treatment groups at month 2, 3 and 4. Scale Bar: 100 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001, n = 5–10.</p>
Full article ">Figure 2 Cont.
<p>Immunohistochemical staining of myofibers (myosin heavy chain expression) in different groups at month 2, 3 and 4 post treatment. (<b>A</b>) Type I muscle fibers (blue), type IIa muscle fibers (green) and type IIb muscle fibers (red) of gastrocnemius at month 2, 3 and 4. (<b>B</b>–<b>D</b>) Type I, IIa and IIb CSA and percentage area in different treatment groups at month 2, 3 and 4. Scale Bar: 100 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001, n = 5–10.</p>
Full article ">Figure 3
<p>Muscle strength and function of SAMP8 mice upon different time points in treatment groups (n = 5). (<b>A</b>) twitch force, (<b>B</b>) specific twitch force, (<b>C</b>) tetanic force and (<b>D</b>) specific twitch force of VIB group were significantly higher than in all other three groups at month 3 post treatment, while the Mg group presented dominantly higher muscle twitch and tetanic contraction parameters at month 4. No significant difference was found at month 2. (<b>E</b>) Grip strength of the Mg group was significantly higher than that of the other groups at month 3, and the Mg + VIB group presented significantly higher grip strength at month 4. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001. (<b>F</b>) Mg treatment and combination treatment showed a higher fatigue curve and significantly higher fatigue force value than the Con group at every time point at month 4. The Mg group compared with Con group: **** <span class="html-italic">p</span> &lt; 0.0001. The VIB group compared with the Con group: ▴ <span class="html-italic">p</span> &lt; 0.05, ▴▴ <span class="html-italic">p</span> &lt; 0.01 and the Mg + VIB group compared with the Con group: # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3 Cont.
<p>Muscle strength and function of SAMP8 mice upon different time points in treatment groups (n = 5). (<b>A</b>) twitch force, (<b>B</b>) specific twitch force, (<b>C</b>) tetanic force and (<b>D</b>) specific twitch force of VIB group were significantly higher than in all other three groups at month 3 post treatment, while the Mg group presented dominantly higher muscle twitch and tetanic contraction parameters at month 4. No significant difference was found at month 2. (<b>E</b>) Grip strength of the Mg group was significantly higher than that of the other groups at month 3, and the Mg + VIB group presented significantly higher grip strength at month 4. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001. (<b>F</b>) Mg treatment and combination treatment showed a higher fatigue curve and significantly higher fatigue force value than the Con group at every time point at month 4. The Mg group compared with Con group: **** <span class="html-italic">p</span> &lt; 0.0001. The VIB group compared with the Con group: ▴ <span class="html-italic">p</span> &lt; 0.05, ▴▴ <span class="html-italic">p</span> &lt; 0.01 and the Mg + VIB group compared with the Con group: # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Immunohistochemical staining of inflammatory markers and M1/M2 macrophage in different groups at month 4 post treatment. White arrow showed the positive area. (<b>A</b>) At month 10, the Mg group showed a significantly lower positive area of C/EBPα staining area than the Con group (<span class="html-italic">p</span> &lt; 0.01). The C/EBPα positive area in the VIB group and the Mg + VIB group were lower than in the Con group, without significance. (<b>B</b>) CD206 positive areas in all treatment groups were significantly lower than in the Con group. (<b>C</b>) The F4/80 positive area showed no significant changes among all groups. (<b>D</b>) VIB, Mg and Mg + VIB groups showed significantly lower LYVE1 positive areas than the Con group at month 10. (<b>E</b>) PAX7-positive cell numbers in the VIB and Mg groups were significantly higher than in the Con group. (<b>F</b>) Quantification results of C/EBPα, CD206, F4/80, LYVE1 positive area and PAX7-positive cell number. Scale Bar: 10 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001, n = 5–10.</p>
Full article ">Figure 4 Cont.
<p>Immunohistochemical staining of inflammatory markers and M1/M2 macrophage in different groups at month 4 post treatment. White arrow showed the positive area. (<b>A</b>) At month 10, the Mg group showed a significantly lower positive area of C/EBPα staining area than the Con group (<span class="html-italic">p</span> &lt; 0.01). The C/EBPα positive area in the VIB group and the Mg + VIB group were lower than in the Con group, without significance. (<b>B</b>) CD206 positive areas in all treatment groups were significantly lower than in the Con group. (<b>C</b>) The F4/80 positive area showed no significant changes among all groups. (<b>D</b>) VIB, Mg and Mg + VIB groups showed significantly lower LYVE1 positive areas than the Con group at month 10. (<b>E</b>) PAX7-positive cell numbers in the VIB and Mg groups were significantly higher than in the Con group. (<b>F</b>) Quantification results of C/EBPα, CD206, F4/80, LYVE1 positive area and PAX7-positive cell number. Scale Bar: 10 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001, n = 5–10.</p>
Full article ">Figure 5
<p>Treatment effects on muscle hypertrophy and atrophy in SAMP8 mice upon different time points. (<b>A</b>–<b>C</b>) Expression of IGF-1, MyoD, MyoG, Myf5 and Myf6 mRNA in different treatment groups at month 2, 3 and 4. (<b>D</b>) MuRF1 and MAFbx mRNA expression levels in the VIB group were significantly lower than in the Con group at month 4. (<b>E</b>,<b>F</b>) Western blots of protein expressions of mTOR, PI3K-p85, Akt, pAkt, p70-S6K, EIF4EBP1, MyoD and GAPDH at month 2, 3 and 4. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001, n = 3.</p>
Full article ">Figure 5 Cont.
<p>Treatment effects on muscle hypertrophy and atrophy in SAMP8 mice upon different time points. (<b>A</b>–<b>C</b>) Expression of IGF-1, MyoD, MyoG, Myf5 and Myf6 mRNA in different treatment groups at month 2, 3 and 4. (<b>D</b>) MuRF1 and MAFbx mRNA expression levels in the VIB group were significantly lower than in the Con group at month 4. (<b>E</b>,<b>F</b>) Western blots of protein expressions of mTOR, PI3K-p85, Akt, pAkt, p70-S6K, EIF4EBP1, MyoD and GAPDH at month 2, 3 and 4. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001, n = 3.</p>
Full article ">Figure 6
<p>Myoblast differentiation and myotube formation in the C2C12 cell line with different treatments with PI3k/Akt inhibitor (LY294002) and mTOR inhibitor (Rapamycin). (<b>A</b>) Immunofluorescent staining of myotubes of VIB, Mg and combined treatment groups appeared morphologically distinct from the Con group. (<b>B</b>) Myotube diameters, myotube nuclei numbers of VIB, Mg and Mg + VIB groups showed a significant difference compared with the Con group. (<b>C</b>,<b>D</b>) Inhibitor groups showed significant smaller myotube diameters and myotube nuclei numbers. Scale Bar: 50 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001, n = 10–15.</p>
Full article ">Figure 6 Cont.
<p>Myoblast differentiation and myotube formation in the C2C12 cell line with different treatments with PI3k/Akt inhibitor (LY294002) and mTOR inhibitor (Rapamycin). (<b>A</b>) Immunofluorescent staining of myotubes of VIB, Mg and combined treatment groups appeared morphologically distinct from the Con group. (<b>B</b>) Myotube diameters, myotube nuclei numbers of VIB, Mg and Mg + VIB groups showed a significant difference compared with the Con group. (<b>C</b>,<b>D</b>) Inhibitor groups showed significant smaller myotube diameters and myotube nuclei numbers. Scale Bar: 50 μm. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001, n = 10–15.</p>
Full article ">Figure 7
<p>Treatment effects on myogenesis, atrogenes mRNA expression and PI3K/Akt/mTOR pathway with inhibitors in the C2C12 myoblast cell line. (<b>A</b>) Heatmap of MRF and atrogenes showed that (<b>B</b>) both the VIB and Mg groups could significantly increase the relative expression level of Myf5 mRNA compared with the Con group. The VIB and Mg + VIB groups showed a similar significantly increasing effect on Myf6 mRNA expression compared with the Con group. (<b>C</b>) Inhibitor groups had significantly decreased Myf5 and MyoG expression. (<b>D</b>,<b>E</b>) Western blots of protein expressions of mTOR, PI3K-p85, Akt, pAkt and GAPDH in different treatment groups in vitro. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, n = 3–5.</p>
Full article ">Figure 7 Cont.
<p>Treatment effects on myogenesis, atrogenes mRNA expression and PI3K/Akt/mTOR pathway with inhibitors in the C2C12 myoblast cell line. (<b>A</b>) Heatmap of MRF and atrogenes showed that (<b>B</b>) both the VIB and Mg groups could significantly increase the relative expression level of Myf5 mRNA compared with the Con group. The VIB and Mg + VIB groups showed a similar significantly increasing effect on Myf6 mRNA expression compared with the Con group. (<b>C</b>) Inhibitor groups had significantly decreased Myf5 and MyoG expression. (<b>D</b>,<b>E</b>) Western blots of protein expressions of mTOR, PI3K-p85, Akt, pAkt and GAPDH in different treatment groups in vitro. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, n = 3–5.</p>
Full article ">
14 pages, 2380 KiB  
Article
Role of Glucocorticoid Signaling and HDAC4 Activation in Diaphragm and Gastrocnemius Proteolytic Activity in Septic Rats
by Álvaro Moreno-Rupérez, Teresa Priego, María Ángeles González-Nicolás, Asunción López-Calderón, Alberto Lázaro and Ana Isabel Martín
Int. J. Mol. Sci. 2022, 23(7), 3641; https://doi.org/10.3390/ijms23073641 - 26 Mar 2022
Cited by 8 | Viewed by 3086
Abstract
Sepsis increases glucocorticoid and decreases IGF-1, leading to skeletal muscle wasting and cachexia. Muscle atrophy mainly takes place in locomotor muscles rather than in respiratory ones. Our study aimed to elucidate the mechanism responsible for this difference in muscle proteolysis, focusing on local [...] Read more.
Sepsis increases glucocorticoid and decreases IGF-1, leading to skeletal muscle wasting and cachexia. Muscle atrophy mainly takes place in locomotor muscles rather than in respiratory ones. Our study aimed to elucidate the mechanism responsible for this difference in muscle proteolysis, focusing on local inflammation and IGF-1 as well as on their glucocorticoid response and HDAC4-myogenin activation. Sepsis was induced in adult male rats by lipopolysaccharide (LPS) injection (10 mg/kg), and 24 h afterwards, rats were euthanized. LPS increased TNFα and IL-10 expression in both muscles studied, the diaphragm and gastrocnemius, whereas IL-6 and SOCS3 mRNA increased only in diaphragm. In comparison with gastrocnemius, diaphragm showed a lower increase in proteolytic marker expression (atrogin-1 and LC3b) and in LC3b protein lipidation after LPS administration. LPS increased the expression of glucocorticoid induced factors, KLF15 and REDD1, and decreased that of IGF-1 in gastrocnemius but not in the diaphragm. In addition, an increase in HDAC4 and myogenin expression was induced by LPS in gastrocnemius, but not in the diaphragm. In conclusion, the lower activation of both glucocorticoid signaling and HDAC4-myogenin pathways by sepsis can be one of the causes of lower sepsis-induced proteolysis in the diaphragm compared to gastrocnemius. Full article
(This article belongs to the Special Issue Glucocorticoid Signaling Pathway: From Bench to Bedside)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of sepsis induced by lipopolysaccharide (LPS) injection (10 mg/kg ip.) on (<b>A</b>) Atrogin-1 mRNA, (<b>B</b>) MuRF1 mRNA, (<b>C</b>) LC3b mRNA, and (<b>D</b>) LC3b-I, and its lipidated form LC3b-II, in rat diaphragm and gastrocnemius muscles, 24 h after a single injection of LPS or saline solution. mRNA was measured by PCR and proteins by Western blot. Representative Western blots are shown in D (bottom). Data represent mean ± standard error of the mean (SEM) for n = 7–8 rats/group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus their respective control rats treated with saline, # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus the diaphragm of rats treated with LPS. Diaph: diaphragm, Gast: gastrocnemius.</p>
Full article ">Figure 2
<p>Effect of sepsis induced by LPS injection (10 mg/kg ip.) on (<b>A</b>) TNFα, (<b>B</b>) IL-10, (<b>C</b>) IL-6 and (<b>D</b>) SOCS3 mRNA, in rat diaphragm and gastrocnemius muscles, 24 h after a single injection of LPS or saline solution. mRNA was measured by PCR. Data represent mean ± standard error of the mean (SEM) for <span class="html-italic">n</span> = 7–8 rats/group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus their respective control rats treated with saline, # <span class="html-italic">p</span> &lt; 0.05 versus diaphragm of rats treated with LPS. Diaph: diaphragm, Gast: gastrocnemius.</p>
Full article ">Figure 3
<p>Effect of sepsis induced by LPS injection (10 mg/kg ip.) on (<b>A</b>) GR, (<b>B</b>) KLF15 and (<b>C</b>) REDD1 mRNA, in rat diaphragm and gastrocnemius muscles, 24 h after a single injection of LPS or saline solution. mRNA was measured by PCR. The data represent mean ± standard error of the mean (SEM) for <span class="html-italic">n</span> = 7–8 rats/group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus their respective control rats treated with saline, # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus diaphragm of rats treated with LPS. Diaph: diaphragm, Gast: gastrocnemius.</p>
Full article ">Figure 4
<p>Effect of sepsis induced by LPS injection (10 mg/kg ip.) on (<b>A</b>) IGF-1R, (<b>B</b>) IGF-1 Ea (IGF-1), and (<b>C</b>) IGFBP-3 mRNA, in rat diaphragm and gastrocnemius muscles, 24 h after a single injection of LPS or saline solution. mRNA was measured by PCR. The data represent mean ± standard error of the mean (SEM) for n = 7–8 rats/group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus their respective control rats treated with saline, ## <span class="html-italic">p</span> &lt; 0.01 versus diaphragm of rats treated with LPS. Diaph: diaphragm, Gast: gastrocnemius.</p>
Full article ">Figure 5
<p>Effect of sepsis induced by LPS injection (10 mg/kg ip.) on (<b>A</b>) HDAC-4 and (<b>B</b>) Myogenin mRNA, (<b>C</b>) HDAC-4, and (<b>D</b>) Myogenin in rat diaphragm and gastrocnemius muscles, 24 h after a single injection of LPS or saline solution. mRNA was measured by PCR and proteins by Western blot. Representative Western blots are shown in (<b>C</b>,<b>D</b> (bottom)). The data represent mean ± standard error of the mean (SEM) for n = 6–8 rats/group. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus their respective control rats treated with saline, # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus diaphragm of rats treated with LPS. Diaph: diaphragm, Gast: gastrocnemius.</p>
Full article ">
17 pages, 2145 KiB  
Article
Exercise Preconditioning Blunts Early Atrogenes Expression and Atrophy in Gastrocnemius Muscle of Hindlimb Unloaded Mice
by Lorenza Brocca, Maira Rossi, Monica Canepari, Roberto Bottinelli and Maria Antonietta Pellegrino
Int. J. Mol. Sci. 2022, 23(1), 148; https://doi.org/10.3390/ijms23010148 - 23 Dec 2021
Cited by 6 | Viewed by 3471
Abstract
A large set of FoxOs-dependent genes play a primary role in controlling muscle mass during hindlimb unloading. Mitochondrial dysfunction can modulate such a process. We hypothesized that endurance exercise before disuse can protect against disuse-induced muscle atrophy by enhancing peroxisome proliferator-activated receptor-γ coactivator-1α [...] Read more.
A large set of FoxOs-dependent genes play a primary role in controlling muscle mass during hindlimb unloading. Mitochondrial dysfunction can modulate such a process. We hypothesized that endurance exercise before disuse can protect against disuse-induced muscle atrophy by enhancing peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) expression and preventing mitochondrial dysfunction and energy-sensing AMP-activated protein kinase (AMPK) activation. We studied cross sectional area (CSA) of muscle fibers of gastrocnemius muscle by histochemistry following 1, 3, 7, and 14 days of hindlimb unloading (HU). We used Western blotting and qRT-PCR to study mitochondrial dynamics and FoxOs-dependent atrogenes’ expression at 1 and 3 days after HU. Preconditioned animals were submitted to moderate treadmill exercise for 7 days before disuse. Exercise preconditioning protected the gastrocnemius from disuse atrophy until 7 days of HU. It blunted alterations in mitochondrial dynamics up to 3 days after HU and the expression of most atrogenes at 1 day after disuse. In preconditioned mice, the activation of atrogenes resumed 3 days after HU when mitochondrial dynamics, assessed by profusion and pro-fission markers (mitofusin 1, MFN1, mitofusin 2, MFN2, optic atrophy 1, OPA1, dynamin related protein 1, DRP1 and fission 1, FIS1), PGC1α levels, and AMPK activation were at a basal level. Therefore, the normalization of mitochondrial dynamics and function was not sufficient to prevent atrogenes activation just a few days after HU. The time course of sirtuin 1 (SIRT1) expression and content paralleled the time course of atrogenes’ expression. In conclusion, seven days of endurance exercise counteracted alterations of mitochondrial dynamics and the activation of atrogenes early into disuse. Despite the normalization of mitochondrial dynamics, the effect on atrogenes’ suppression died away within 3 days of HU. Interestingly, muscle protection lasted until 7 days of HU. A longer or more intense exercise preconditioning may prolong atrogenes suppression and muscle protection. Full article
Show Figures

Figure 1

Figure 1
<p>A single exercise bout and 7-day endurance training increase mRNA and protein expression of PGC 1α in ground <span class="html-italic">mice</span>. Gene expression and protein content analysis of PGC 1α (<b>a</b>) and activation level of AMP-kinase (<b>b</b>) in gastrocnemius of <span class="html-italic">mice</span> subjected to a single bout of treadmill exercise and relative representative Western blot; protein levels were normalized for tubulin expression. Ground: Control <span class="html-italic">mice</span>; Ground SB-EX + 0 h: <span class="html-italic">Mice</span> subjected to a single bout of exercise sacrificed immediately or 3 h after exercise (Ground SB-EX + 3 h). Gene expression and protein content analysis of PGC 1α (<b>c</b>) and activation level of AMP-kinase (<b>d</b>) in gastrocnemius of trained <span class="html-italic">mice</span> and relative representative Western blot; protein levels were normalized for tubulin expression. Ground: Control <span class="html-italic">mice</span>; Ground EX7 + 3 h: <span class="html-italic">Mice</span> subjected to 7 days of endurance exercise sacrificed 3 h after the last section of exercise. The activation level of AMP-kinase was determined through the ratio between the content of the phosphorylated (<span class="html-italic">p</span>) and total form. Data are presented as means ± SD * <span class="html-italic">p</span> &lt; 0.05 *** <span class="html-italic">p</span> &lt; 0.0005 **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Physical preconditioning mitigates disuse-induced atrophy of gastrocnemius. Cross Sectional Area (CSA) of gastrocnemius muscle fibers of <span class="html-italic">mice</span> subjected to disuse alone and preceded by physical preconditioning and representative hematoxylin–eosin staining; scale bars: 100µm (<b>a</b>). CSA time course changes with disuse alone and preceded by 7 days of physical preconditioning (<b>b</b>). Ground: Control <span class="html-italic">mice</span>; HU1, HU3, HU7, and HU14: <span class="html-italic">Mice</span> subjected to 1, 3, 7, and 14 days of hindlimb unloading; Ground EX7: <span class="html-italic">Mice</span> subjected to 7 days of physical exercise. Data are presented as means ± SD * <span class="html-italic">p</span> &lt; 0.05 ** <span class="html-italic">p</span> &lt; 0.005 *** <span class="html-italic">p</span> &lt; 0.0005. §: Different from ground and unexercised HU1 groups; †: Different from preconditioned HU3 group; ∗: Different from ground and preconditioned HU1 and HU3 groups.</p>
Full article ">Figure 3
<p>Physical preconditioning counteracts the early atrogenes’ induction during disuse. RT-PCR analysis of transcriptional levels of <span class="html-italic">FoxO3</span> and the muscle-specific ubiquitin ligases <span class="html-italic">MuRF1</span>, <span class="html-italic">Atrogin1</span>, <span class="html-italic">SMART</span>, <span class="html-italic">MUSA1</span>, <span class="html-italic">FbxO31</span>, proteasome subunit atrogenes (<span class="html-italic">Psmd11, Psme4</span>), de-ubiquitinating enzyme atrogene (<span class="html-italic">USP14</span>), ubiquitin atrogene (<span class="html-italic">UBC</span>), autophagy-related atrogenes (<span class="html-italic">Cathepsin l, P62, BNIP3</span>), AMP deaminase atrogene (<span class="html-italic">Ampd3</span>), oxidative stress related atrogene (<span class="html-italic">Mt1</span>), unfolded protein response atrogene (<span class="html-italic">Gadd34</span>), transcription regulators Smad2/3 atrogene (<span class="html-italic">TGIF</span>). The expression levels were normalized on <span class="html-italic">GAPDH</span>. Ground: Control <span class="html-italic">mice</span>; HU1: <span class="html-italic">Mice</span> subjected to 1 day of hindlimb unloading; EX7 + HU1: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 1 day of hindlimb unloading; HU3: <span class="html-italic">Mice</span> subjected to 3 days of hindlimb unloading; EX7 + HU3: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 3 days of hindlimb unloading. Data are presented as means ± SD * <span class="html-italic">p</span> &lt; 0.05 ** <span class="html-italic">p</span> &lt; 0.005 *** <span class="html-italic">p</span> &lt; 0.0005 **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Physical preconditioning effect on phosphorylation of kinases of the Akt/mTOR pathway during disuse. Determination of activity levels of <span class="html-italic">p</span>-AKT (<b>a</b>), <span class="html-italic">p</span>-mTOR (<b>b</b>), and <span class="html-italic">p</span>-4EBP1 (<b>c</b>) by Western blot analysis of the ratio between the content in the phosphorylated (<span class="html-italic">p</span>) and total forms and relative representative Western blot. Ground: Control <span class="html-italic">mice</span>; HU1: <span class="html-italic">Mice</span> subjected to 1 day of hindlimb unloading; EX7 + HU1: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 1 day of hindlimb unloading; HU3: <span class="html-italic">Mice</span> subjected to 3 days of hindlimb unloading; EX7 + HU3: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 3 days of hindlimb unloading. Data are presented as means ± SD * <span class="html-italic">p</span> &lt; 0.05 ** <span class="html-italic">p</span> &lt; 0.005 *** <span class="html-italic">p</span> &lt; 0.0005 **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Physical preconditioning counteracts the decrease in MFN2 pro-fusion protein induced by disuse. Quantification of protein content of mitochondria dynamics markers by Western blotting. Pro-fusion proteins MFN1 (<b>a</b>), MFN2 (<b>b</b>), OPA1 (<b>c</b>), pro-fission proteins <span class="html-italic">p</span>-DRP1<sub>(Ser616)</sub> (<b>d</b>), <span class="html-italic">p</span>-DRP1<sub>(Ser637)</sub> (<b>e</b>), and FIS1 (<b>f</b>), and relative representative Western blot. Ground: Control <span class="html-italic">mice</span>; HU1: <span class="html-italic">Mice</span> subjected to 1 day of hindlimb unloading; EX7 + HU1: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 1 day of hindlimb unloading; HU3: <span class="html-italic">Mice</span> subjected to 3 days of hindlimb unloading; EX7 + HU3: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 3 days of hindlimb unloading. Protein levels were normalized for tubulin expression. The phosphorylation status of DRP1<sub>(Ser616)</sub> and DRP1<sub>(Ser637)</sub> was determined through the ratio between the content in the phosphorylated and total forms. Data are presented as means ± SD * <span class="html-italic">p</span> &lt; 0.05 ** <span class="html-italic">p</span> &lt; 0.005 *** <span class="html-italic">p</span> &lt; 0.0005.</p>
Full article ">Figure 6
<p>Effects of hindlimb unloading and exercise preconditioning on FoxO regulators. Activation level of AMP-kinase (<b>a</b>), Gene expression and protein content analysis of PGC1α (<b>b</b>), gene expression and protein content analysis of SIRT1 (<b>c</b>), and relative representative Western blot. Ground: Control <span class="html-italic">mice</span>; HU1: <span class="html-italic">Mice</span> subjected to 1 day of hindlimb unloading; EX7 + HU1: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 1 day of hindlimb unloading; HU3: <span class="html-italic">Mice</span> subjected to 3 days of hindlimb unloading; EX7 + HU3: <span class="html-italic">Mice</span> subjected to 7 days of physical preconditioning followed by 3 days of hindlimb unloading. Data are presented as means ± SD * <span class="html-italic">p</span> &lt; 0.05 ** <span class="html-italic">p</span> &lt; 0.005 *** <span class="html-italic">p</span> &lt; 0.0005 **** <span class="html-italic">p</span> &lt; 0.0001. Gene expression levels were determined by RT-PCR analysis and normalized on <span class="html-italic">GAPDH</span>. Protein content levels were determined by Western blot analysis and normalized for tubulin. The activation level of AMP-kinase was determined by Western blot analysis of the ratio between the content of the phosphorylated and total form.</p>
Full article ">
9 pages, 871 KiB  
Review
The Novel Role of PGC1α in Bone Metabolism
by Cinzia Buccoliero, Manuela Dicarlo, Patrizia Pignataro, Francesco Gaccione, Silvia Colucci, Graziana Colaianni and Maria Grano
Int. J. Mol. Sci. 2021, 22(9), 4670; https://doi.org/10.3390/ijms22094670 - 28 Apr 2021
Cited by 18 | Viewed by 3282
Abstract
Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) is a protein that promotes transcription of numerous genes, particularly those responsible for the regulation of mitochondrial biogenesis. Evidence for a key role of PGC1α in bone metabolism is very recent. In vivo studies showed that [...] Read more.
Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) is a protein that promotes transcription of numerous genes, particularly those responsible for the regulation of mitochondrial biogenesis. Evidence for a key role of PGC1α in bone metabolism is very recent. In vivo studies showed that PGC1α deletion negatively affects cortical thickness, trabecular organization and resistance to flexion, resulting in increased risk of fracture. Furthermore, in a mouse model of bone disease, PGC1α activation stimulates osteoblastic gene expression and inhibits atrogene transcription. PGC1α overexpression positively affects the activity of Sirtuin 3, a mitochondrial nicotinammide adenina dinucleotide (NAD)-dependent deacetylase, on osteoblastic differentiation. In vitro, PGC1α overexpression prevents the reduction of mitochondrial density, membrane potential and alkaline phosphatase activity caused by Sirtuin 3 knockdown in osteoblasts. Moreover, PGC1α influences the commitment of skeletal stem cells towards an osteogenic lineage, while negatively affects marrow adipose tissue accumulation. In this review, we will focus on recent findings about PGC1α action on bone metabolism, in vivo and in vitro, and in pathologies that cause bone loss, such as osteoporosis and type 2 diabetes. Full article
Show Figures

Figure 1

Figure 1
<p>PGC1α deletion affects bone. (<b>A</b>) Representative images of micro-CT-generated sections of the tibia midshaft of PGC1α+/+ and PGC1α−/− mice show a reduction in cortical thickness (Ct. Th) in the absence of PGC1α. Adapted from [<a href="#B12-ijms-22-04670" class="html-bibr">12</a>]. (<b>B</b>) Schematic representation of the effect of PGC1α deletion in osteoblasts consisting of the reduction of <span class="html-italic">Ocn</span> and <span class="html-italic">Col1a1</span> levels.</p>
Full article ">Figure 2
<p>PGC1α deletion affects marrow adipose tissue (MAT). (<b>A</b>) Photomicrographs of hematoxylin and eosin-stained sections of MAT from PGC1α +/+ and PGC1α −/− (magnification: 20×) show an increased number of adipocytes in the absence of PGC1α <span class="html-italic">(unpublished data)</span>. (<b>B</b>) Schematic representation of the effect of PGC1α deletion in bone marrow adipocytes consisting of the increase of <span class="html-italic">C/EBPα</span> expression.</p>
Full article ">
36 pages, 2900 KiB  
Review
Ubiquitin Ligases at the Heart of Skeletal Muscle Atrophy Control
by Dulce Peris-Moreno, Laura Cussonneau, Lydie Combaret, Cécile Polge and Daniel Taillandier
Molecules 2021, 26(2), 407; https://doi.org/10.3390/molecules26020407 - 14 Jan 2021
Cited by 39 | Viewed by 7122
Abstract
Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a [...] Read more.
Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a master regulator of skeletal muscle that impacts muscle contractile properties and metabolism through multiple levers like signaling pathways, contractile apparatus degradation, etc. Among the different actors of the UPS, the E3 ubiquitin ligases specifically target key proteins for either degradation or activity modulation, thus controlling both pro-anabolic or pro-catabolic factors. The atrogenes MuRF1/TRIM63 and MAFbx/Atrogin-1 encode for key E3 ligases that target contractile proteins and key actors of protein synthesis respectively. However, several other E3 ligases are involved upstream in the atrophy program, from signal transduction control to modulation of energy balance. Controlling E3 ligases activity is thus a tempting approach for preserving muscle mass. While indirect modulation of E3 ligases may prove beneficial in some situations of muscle atrophy, some drugs directly inhibiting their activity have started to appear. This review summarizes the main signaling pathways involved in muscle atrophy and the E3 ligases implicated, but also the molecules potentially usable for future therapies. Full article
Show Figures

Figure 1

Figure 1
<p>Signaling pathways regulating skeletal muscle mass and function. Myofiber representation of the different signaling pathways controlling skeletal muscle mass and function during atrophy conditions. Ligands and arrows (both with head or perpendicular line) in green denote those signaling pathways and interactions with an anabolic effect whereas the red ones represent catabolic signaling. Orange ligands and arrows stand for pathways with a dual role (context-dependent). ß2-AR: ß-2 Adrenergic Receptor; <math display="inline"><semantics> <mi>γ</mi> </semantics></math>-sec: <math display="inline"><semantics> <mi>γ</mi> </semantics></math>-secretase; Ang: Angiotensin; AT1R: Angiotensin II Type 1 Receptor; AT2R: Angiotensin II Type 2 Receptor; BCAAs: Branched-chain amino acids; BMP R: Bone Morphogenetic Receptor; Calp: Calpain; CSL: CBF1, Suppressor of Hairless, Lag-1; Dsh: Dishevelled; Fzd: Frizzled; GR: Glucocorticoid Receptor; IL-6: Interleukin-6; NCID: Notch Intracellular domain; NOX: NADPH oxidase activator; P: Phosphorylation; S1P: Sphingosine-1-phosphate; SLC T: Solute Carrier Transporter; STAPs: Signal Transducing Adaptor Proteins; TF: Transcription Factors; TGF-ß R: Transforming Growth Factor ß Receptor; TKR: Tyrosine-protein Kinase Receptor; TLR: Toll-like Receptor; TNF R: Tumor Necrosis Factor Receptor; Transl. Fact.: Translational Factors.</p>
Full article ">Figure 2
<p>E3 ubiquitin ligases regulating skeletal muscle mass and molecules developed to modulate their activity and expression. Myofiber representation of the different E3-ligases and molecules targeting the signaling pathways controlling skeletal muscle mass and function during atrophy conditions. Ligands and arrows (both with head or perpendicular line) in green denote those signaling pathways and interactions with an anabolic effect whereas the red ones indicate catabolic signaling. ß2-AR: ß-2 Adrenergic Receptor; BCAAs: Branched-chain amino acids; BMP R: Bone Morphogenetic Receptor; Calp: Calpain; CSL: CBF1, Suppressor of Hairless, Lag-1; GR: Glucocorticoid Receptor; IL-6: Interleukin-6; NCID: Notch Intracellular domain; NOX: NADPH oxidase activator; P: Phosphorylation; STAPs: Signal Transducing Adaptor Proteins; TF: Transcription Factors; TGF-ß R: Transforming Growth Factor ß Receptor; TKR: Tyrosine-protein Kinase Receptor; TLR: Toll-like Receptor; TNF R: Tumor Necrosis Factor Receptor; Transl. Fact.: Translational Factors.</p>
Full article ">Figure 2 Cont.
<p>E3 ubiquitin ligases regulating skeletal muscle mass and molecules developed to modulate their activity and expression. Myofiber representation of the different E3-ligases and molecules targeting the signaling pathways controlling skeletal muscle mass and function during atrophy conditions. Ligands and arrows (both with head or perpendicular line) in green denote those signaling pathways and interactions with an anabolic effect whereas the red ones indicate catabolic signaling. ß2-AR: ß-2 Adrenergic Receptor; BCAAs: Branched-chain amino acids; BMP R: Bone Morphogenetic Receptor; Calp: Calpain; CSL: CBF1, Suppressor of Hairless, Lag-1; GR: Glucocorticoid Receptor; IL-6: Interleukin-6; NCID: Notch Intracellular domain; NOX: NADPH oxidase activator; P: Phosphorylation; STAPs: Signal Transducing Adaptor Proteins; TF: Transcription Factors; TGF-ß R: Transforming Growth Factor ß Receptor; TKR: Tyrosine-protein Kinase Receptor; TLR: Toll-like Receptor; TNF R: Tumor Necrosis Factor Receptor; Transl. Fact.: Translational Factors.</p>
Full article ">
14 pages, 1622 KiB  
Article
Phytoecdysteroids Do Not Have Anabolic Effects in Skeletal Muscle in Sedentary Aging Mice
by Marcus M. Lawrence, Kevin A. Zwetsloot, Susan T. Arthur, Chase A. Sherman, Joshua R. Huot, Vladimir Badmaev, Mary Grace, Mary Ann Lila, David C. Nieman and R. Andrew Shanely
Int. J. Environ. Res. Public Health 2021, 18(2), 370; https://doi.org/10.3390/ijerph18020370 - 6 Jan 2021
Cited by 6 | Viewed by 4644
Abstract
Skeletal muscle mass and strength are lost with aging. Phytoecdysteroids, in particular 20-hydroxyecdysone (20E), increase protein synthesis in C2C12 skeletal muscle cells and muscle strength in young rats. The objective of this study was to determine whether an extract from Ajuga turkestanica (ATE), [...] Read more.
Skeletal muscle mass and strength are lost with aging. Phytoecdysteroids, in particular 20-hydroxyecdysone (20E), increase protein synthesis in C2C12 skeletal muscle cells and muscle strength in young rats. The objective of this study was to determine whether an extract from Ajuga turkestanica (ATE), enriched in phytoecdysteroids, and 20E affect skeletal muscle mass and fiber size, fiber type, activation of the PI3K–Akt signaling pathway, and the mRNA levels of MAFbx, MuRF-1, and myostatin in sedentary aging mice. Aging male C57BL/6 mice (20 months old) received ATE, 20E, or vehicle (CT) once per day for 28 days or a single acute dose. Treatment did not alter body, muscle, or organ mass; fiber cross-sectional area; or fiber type in the triceps brachii or plantaris muscles. Likewise, protein synthesis signaling markers (i.e., phosphorylation of AktSer473 and p70S6kThr389) measured after either 28 days or acutely were unchanged. Neither ATE nor 20E treatment for 28 days affected the mRNA levels of MAFbx, MuRF-1, and myostatin. In conclusion, these data indicate that phytoecdysteroid treatment does not alter muscle mass or fiber type, nor does it activate protein synthesis signaling in the skeletal muscle of sedentary aging mice. Full article
(This article belongs to the Special Issue Regulation of Muscle Mass, Exercise, Metabolism)
Show Figures

Figure 1

Figure 1
<p>Immunohistochemical analysis of muscle fiber type. Representative images of fiber type and laminin staining on entire plantaris and triceps brachii cross-sections. Blue, type IIa; unstained/black, type IIx; green, type IIb; red, laminin. Scale bar = 200 μm.</p>
Full article ">Figure 2
<p>Protein analysis of markers of protein synthesis signaling in aging gastrocnemius muscle via Western blot after the 28-day supplementation period. Quantification of phosphorylated Akt (<b>A</b>), total Akt protein (<b>B</b>), phosphorylated p70S6K (<b>C</b>), and total p70S6K protein (<b>D</b>) by treatment, respectively. Representative Western blot protein bands are provided. Data are normalized to CT and presented as mean ± SEM. CT, control; ATE, <span class="html-italic">A. turkestanica</span> extract; 20E, 20-hydroxyecdysone. No significant differences were found, <span class="html-italic">p</span> &gt; 0.05, all.</p>
Full article ">Figure 3
<p>Protein analysis of markers of protein synthesis signaling in aging gastrocnemius muscle via Western blot after an acute dose of supplementation. Quantification of phosphorylated Akt (<b>A</b>), phosphorylated p70S6K (<b>B</b>), phosphorylated 4EBP-1 (<b>C</b>), and phosphorylated rpS6 (<b>D</b>) protein by treatment, respectively. Representative Western blot protein bands are provided. Data are normalized to CT and presented as mean ± SEM. CT, control; ATE, <span class="html-italic">A. turkestanica</span> extract; 20E, 20-hydroxyecdysone. No significant differences were found, <span class="html-italic">p</span> &gt; 0.05, all.</p>
Full article ">Figure 4
<p>Analysis of mRNA levels for the atrogenes, <span class="html-italic">MAFBx</span> and <span class="html-italic">MuRF-1</span>, and the negative regulator of muscle mass, <span class="html-italic">myostatin</span> in aging gastrocnemius muscle via real-time PCR after the 28-day supplementation period. Quantification of <span class="html-italic">MAFBx</span> (<b>A</b>), <span class="html-italic">MuRF-1</span> (<b>B</b>), and <span class="html-italic">myostatin</span> (<b>C</b>) mRNA levels by treatment, respectively. Data are normalized to CT and presented as mean ± SEM. CT, control; ATE, <span class="html-italic">A. turkestanica</span> extract; 20E, 20-hydroxyecdysone. No significant differences were found, <span class="html-italic">p</span> &gt; 0.05, all.</p>
Full article ">
37 pages, 1907 KiB  
Review
Master Regulators of Muscle Atrophy: Role of Costamere Components
by Luisa Gorza, Matteo Sorge, Laura Seclì and Mara Brancaccio
Cells 2021, 10(1), 61; https://doi.org/10.3390/cells10010061 - 3 Jan 2021
Cited by 21 | Viewed by 9356
Abstract
The loss of muscle mass and force characterizes muscle atrophy in several different conditions, which share the expression of atrogenes and the activation of their transcriptional regulators. However, attempts to antagonize muscle atrophy development in different experimental contexts by targeting contributors to the [...] Read more.
The loss of muscle mass and force characterizes muscle atrophy in several different conditions, which share the expression of atrogenes and the activation of their transcriptional regulators. However, attempts to antagonize muscle atrophy development in different experimental contexts by targeting contributors to the atrogene pathway showed partial effects in most cases. Other master regulators might independently contribute to muscle atrophy, as suggested by our recent evidence about the co-requirement of the muscle-specific chaperone protein melusin to inhibit unloading muscle atrophy development. Furthermore, melusin and other muscle mass regulators, such as nNOS, belong to costameres, the macromolecular complexes that connect sarcolemma to myofibrils and to the extracellular matrix, in correspondence with specific sarcomeric sites. Costameres sense a mechanical load and transduce it both as lateral force and biochemical signals. Recent evidence further broadens this classic view, by revealing the crucial participation of costameres in a sarcolemmal “signaling hub” integrating mechanical and humoral stimuli, where mechanical signals are coupled with insulin and/or insulin-like growth factor stimulation to regulate muscle mass. Therefore, this review aims to enucleate available evidence concerning the early involvement of costamere components and additional putative master regulators in the development of major types of muscle atrophy. Full article
(This article belongs to the Special Issue Skeletal Muscle Atrophy: Mechanisms at a Cellular Level)
Show Figures

Figure 1

Figure 1
<p>The neuronal NOSμ isoform interacts with the Grp94/gp96 chaperone and is delivered at the subsarcolemma by docking at the DCG. Unloading-induced mitochondrial ROS production causes nNOSμ untethering from DGC and translocation in the sarcoplasm, where the enzyme through either “coupled” or “uncoupled” NADPH oxidation (inset) leads to NO/O<sub>2</sub><sup>−</sup> production, respectively, and FoxO3 activation. NO = nitric oxide; nNOS = neuronal nitric oxide synthase; SR-ER = sarco-endoplasmic reticulum; IGF1 = insulin-like growth factor 1.</p>
Full article ">Figure 2
<p>The sarcolemmal costamere components and their interactors form a supramolecular platform specialized in mechanostransduction and signal integration (only a part of the components is shown in the figure). ECM = extracellular matrix; ILK = integrin-linked kinase; MLP = muscle LIM protein; FAK = focal adhesion kinase; nNOS = neuronal nitric oxide synthase; PI3K = phosphoinositide 3-kinase IRS-1 = insulin receptor substrate-1; IGF1R =insulin-like growth factor 1 receptor; SR = sarcoplasmic reticulum.</p>
Full article ">Figure 3
<p>Signaling pathways activated after a 6–12 h bout of muscle unloading in costameres. Continuous lines indicate stimulatory effects, while discontinuous lines indicate inhibitory effects. Cbl-b = Casitas B-lineage lymphoma-b ubiquitin ligase; Ub = ubiquitin; nNOS = neuronal nitric oxide synthase; FOXO3 = forkhead box O3; MuRF1 = muscle RING-finger protein-1; MAFbx = muscle atrophy F-box; HDAC1 = histone deacetylase 1; Ac = acetylation; IRS-1 = insulin receptor substrate-1; 70S6K = Ribosomal protein S6 kinase p70; P = phosphorylation; AMPK = AMP-activated protein kinase.</p>
Full article ">
15 pages, 1983 KiB  
Article
Beneficial Effects of a Mixture of Algae and Extra Virgin Olive Oils on the Age-Induced Alterations of Rodent Skeletal Muscle: Role of HDAC-4
by Daniel González-Hedström, Teresa Priego, Asunción López-Calderón, Sara Amor, María de la Fuente-Fernández, Antonio Manuel Inarejos-García, Ángel Luis García-Villalón, Ana Isabel Martín and Miriam Granado
Nutrients 2021, 13(1), 44; https://doi.org/10.3390/nu13010044 - 25 Dec 2020
Cited by 10 | Viewed by 3740
Abstract
Aging is associated with a progressive decline in skeletal muscle mass, strength and function (sarcopenia). We have investigated whether a mixture of algae oil (25%) and extra virgin olive oil (75%) could exert beneficial effects on sarcopenia. Young (3 months) and old (24 [...] Read more.
Aging is associated with a progressive decline in skeletal muscle mass, strength and function (sarcopenia). We have investigated whether a mixture of algae oil (25%) and extra virgin olive oil (75%) could exert beneficial effects on sarcopenia. Young (3 months) and old (24 months) male Wistar rats were treated with vehicle or with the oil mixture (OM) (2.5 mL/kg) for 21 days. Aging decreased gastrocnemius weight, total protein, and myosin heavy chain mRNA. Treatment with the OM prevented these effects. Concomitantly, OM administration decreased the inflammatory state in muscle; it prevented the increase of pro-inflammatory interleukin-6 (IL-6) and the decrease in anti-inflammatory interleukin-10 (IL-10) in aged rats. The OM was not able to prevent aging-induced alterations in either the insulin-like growth factor I/protein kinase B (IGF-I/Akt) pathway or in the increased expression of atrogenes in the gastrocnemius. However, the OM prevented decreased autophagy activity (ratio protein 1A/1B-light chain 3 (LC3b) II/I) induced by aging and increased expression of factors related with muscle senescence such as histone deacetylase 4 (HDAC-4), myogenin, and IGF-I binding protein 5 (IGFBP-5). These data suggest that the beneficial effects of the OM on muscle can be secondary to its anti-inflammatory effect and to the normalization of HDAC-4 and myogenin levels, making this treatment an alternative therapeutic tool for sarcopenia. Full article
(This article belongs to the Special Issue Health Benefits of Edible Oils)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Beneficial effects of oil mixture on body weight gain (<b>A</b>), gastrocnemius muscle relative weight (<b>B</b>), gastrocnemius muscle protein concentration (<b>C</b>), and gastrocnemius mRNA levels of myosin heavy chain isoforms (MHC) I and IIa (<b>D</b>). Values are represented as mean ± standard error of the mean (SEM) of young rats (<span class="html-italic">n</span> = 11), old rats (<span class="html-italic">n</span> = 8), and old rats treated for 21 days with the oil mixture (OLD + OILS, <span class="html-italic">n</span> = 5). Statistics: ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 vs. Young; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. Old, by least-significant difference post hoc analysis after significant one-way analysis of variance (ANOVA).</p>
Full article ">Figure 2
<p>Oil mixture treatment improved the inflammatory profile and the expression of metabolic regulators in old gastrocnemius muscle. Figures show the mRNA levels of (<b>A</b>) interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), IL-1β and interleukin-6 (IL-10), (<b>B</b>) peroxisome proliferator-activated receptor alpha (PPAR-α), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) of young rats (<span class="html-italic">n</span> = 11), old rats (<span class="html-italic">n</span> = 8), and old rats treated for 21 days with the oil mixture (OLD + OILS, <span class="html-italic">n</span> = 5). Values are represented as mean ± standard error of the mean (SEM). Statistics: ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 vs. Young; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. Old, by least-significant difference post hoc analysis after significant one-way analysis of variance (ANOVA).</p>
Full article ">Figure 3
<p>Oil mixture treatment did not prevent the age-induced decrease of IGF-I serum levels and the muscle Akt signaling pathway, although it did prevent increased expression of muscle IGFBP-5. Figures show (<b>A</b>) insulin-like growth factor (IGF-I) serum levels; (<b>B</b>) gastrocnemius IGF-I, IGF-I receptor (IGF-IR), IGF-I binding protein 3 (IGFBP-3), and 5 (IGFBP-5) mRNA levels; (<b>C</b>) gastrocnemius protein levels and ratio between phospho Akt (pAkt) and Akt of young rats (<span class="html-italic">n</span> = 11), old rats (<span class="html-italic">n</span> = 8) and old rats treated for 21 days with the oil mixture (OLD + OILS, <span class="html-italic">n</span> = 5). Values are represented as mean ± standard error of the mean (SEM). Statistics: ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 vs. Young, by least-significant difference post hoc analysis after significant one-way analysis of variance (ANOVA).</p>
Full article ">Figure 4
<p>Oil mixture treatment did not prevent age-induced alterations in the expression of atrogenes and Bnip, although it did prevent a decrease in autophagy activity (ratio LC3b II/I) in skeletal muscle. Figures show gastrocnemius (<b>A</b>) muscle RING-finger protein-1 (MuRF1) and atrogin-1; (<b>B</b>) BCL2/adenovirus E1B 19 kDa interacting protein (Bnip) and 1A/1B-light chain 3 (LC3b) mRNA levels; (<b>C</b>) gastrocnemius protein levels and ratio between LC3b I and II of young rats (<span class="html-italic">n</span> = 11), old rats (<span class="html-italic">n</span> = 8) and old rats treated for 21 days with the oil mixture (OLD + OILS, <span class="html-italic">n</span> = 5). Values are represented as mean ± standard error of the mean (SEM). Statistics: ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 vs. Young; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. Old, by least-significant difference post hoc analysis after significant one-way analysis of variance (ANOVA).</p>
Full article ">Figure 5
<p>Oil mixture treatment prevented the age-induced activation of the HADC-4-myogenin axis in skeletal muscle. Figures show gastrocnemius mRNA levels of (<b>A</b>) histone deacetylase 4 (HDAC-4) and myogenin and (<b>B</b>) HDAC-4 protein levels of young rats (<span class="html-italic">n</span> = 11), old rats (<span class="html-italic">n</span> = 8) and old rats treated for 21 days with the oil mixture (OLD + OILS, <span class="html-italic">n</span> = 5). Values are represented as mean ± standard error of the mean (SEM). Statistics: ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span>&lt; 0.05 vs. Young; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. Old, by least-significant difference post hoc analysis after significant one-way analysis of variance (ANOVA).</p>
Full article ">Figure 6
<p>Schematic representation of the effects of the oil mixture on the main aged-induced alterations in skeletal muscle. Oil mixture administration prevented the age-induced increase of HDAC-4. This effect, together with the decrease in inflammatory mediators, may have beneficial consequences such as the prevention of the fiber senescence and muscle atrophy, restoration of autophagy equilibrium, increase in mitochondriogenesis, fatty acid oxidation rate, and protein synthesis. All these effects help to prevent the development of sarcopenia (see Discussion for further details). Abbreviations: BP-5 (insulin-like growth factor I binding protein 5); HDAC-4 (histone deacetylase 4); IL (interleukin); LC3 (1A/1B-light chain 3); MHC (myosin heavy chain); PGC-1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha); PPAR-α (proliferator-activated receptor alpha).</p>
Full article ">
23 pages, 3169 KiB  
Article
Marked Increased Production of Acute Phase Reactants by Skeletal Muscle during Cancer Cachexia
by Isabelle S. Massart, Geneviève Paulissen, Audrey Loumaye, Pascale Lause, Sarah A. Pötgens, Morgane M. Thibaut, Estelle Balan, Louise Deldicque, Azeddine Atfi, Edouard Louis, Damien Gruson, Laure B. Bindels, Marie-Alice Meuwis and Jean-Paul Thissen
Cancers 2020, 12(11), 3221; https://doi.org/10.3390/cancers12113221 - 31 Oct 2020
Cited by 12 | Viewed by 3760
Abstract
Loss of skeletal muscle mass in cancer cachexia is recognized as a predictor of mortality. This study aimed to characterize the changes in the muscle secretome associated with cancer cachexia to gain a better understanding of the mechanisms involved and to identify secreted [...] Read more.
Loss of skeletal muscle mass in cancer cachexia is recognized as a predictor of mortality. This study aimed to characterize the changes in the muscle secretome associated with cancer cachexia to gain a better understanding of the mechanisms involved and to identify secreted proteins which may reflect this wasting process. The changes in the muscle proteome of the C26 model were investigated by label-free proteomic analysis followed by a bioinformatic analysis in order to identify potentially secreted proteins. Multiple reaction monitoring and Western blotting were used to verify the presence of candidate proteins in the circulation. Our results revealed a marked increased muscular production of several acute phase reactants (APR: Haptoglobin, Serine protease inhibitor A3N, Complement C3, Serum amyloid A-1 protein) which are released in the circulation during C26 cancer cachexia. This was confirmed in other models of cancer cachexia as well as in cancer patients. Glucocorticoids and proinflammatory cytokines are responsible for an increased production of APR by muscle cells. Finally, their muscular expressions are strongly positively correlated with body weight loss as well as the muscular induction of atrogens. Our study demonstrates therefore a marked increased production of APR by the muscle in cancer cachexia. Full article
Show Figures

Figure 1

Figure 1
<p>C26 colon carcinoma causes body weight loss and severe muscle atrophy. (<b>A</b>) Body weight evolution of male mice injected with C26 cells (C26) or vehicle (CT). (<b>B</b>) Tibialis anterior (TA) and gastrocnemius (GC) muscle weights from C26 and CT mice 10 days after injection. (<b>C</b>) Gene expression levels of major atrogens in GC muscle of C26 and CT mice. Data are reported as means ± SEM (<span class="html-italic">n</span> = 8/group). * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 vs CT.</p>
Full article ">Figure 2
<p>C26 cancer-induced cachexia is associated with profound changes in the skeletal muscle proteome. Global and differential proteomic analyses of gastrocnemius (GC) muscle from male mice injected with C26 cells (C26) or vehicle (CT). (<b>A</b>) Total number of confident protein identifications and differentially abundant proteins (DAPs) between C26 and CT muscle (<span class="html-italic">p</span> &lt; 0.05) in the sarcoplasmic (SF) and the myofibrillar (MF) fractions. Venn diagram shows shared and unique significant DAPs in the two fractions. Full protein lists are shown in <a href="#app1-cancers-12-03221" class="html-app">Table S1</a>. DAPs found in the differential analysis are communicated in <a href="#app1-cancers-12-03221" class="html-app">Table S2</a>. (<b>B</b>) Volcano plots of the distribution of the 228 and 196 DAPs in the SF and MF, respectively. Red plots represent the 33 common proteins between the two fractions. Y-axis represents -Log10 <span class="html-italic">p</span>-value and x-axis represents Log2 ratio (C26/CT). The dark line shows the significance threshold limit (<span class="html-italic">p</span> = 0.05). (<b>C</b>) Heatmap of DAPs found in SF and MF in each mouse (<span class="html-italic">n</span> = 6/group) showing downregulated proteins in green and upregulated proteins in red. (<b>D</b>) Volcano plot of the proteins the most dysregulated (in red) in the skeletal muscle of C26 mice. Top: list of proteins only detected in one group (CT only or C26 only) in the SF and the MF. Bottom: DAPs between C26 and CT muscle with y-axis representing −Log10 <span class="html-italic">p</span>-value (threshold limit line at <span class="html-italic">p</span> = 0.05) and x-axis representing Log2 ratio C26/CT with vertical lines at +1 or −1 Log2 ratio (C26/CT) corresponding to a ratio C26/CT = 2 and C26/CT = 0.5, respectively.</p>
Full article ">Figure 3
<p>Marked production of acute phase reactants (APR) is observed in the skeletal muscle of C26 cancer-induced cachexia. (<b>A</b>) Hp, Serpina3n, C3, and SAA1 light/heavy peptide ratio measured by MRM in the plasma of C26 and CT male mice. Graphs represent the total of the two most intense transitions for one representative peptide of each protein (being a peptide without M and with the most intense mean signal observed for each protein in the C26 group). GSFPWQAK: peptide of Hp, GPGGVWAAEK: peptide of SAA1, GVFVLNK: peptide of C3, LINDYVR: peptide of Serpina3n. (<b>B</b>) Hp, Serpina3n, C3, and SAA1 protein levels measured by Western blot in the serum of C26 and CT mice. Coomassie blue staining was used as loading control. Hp, Serpina3n, C3, and SAA1 proteins. Please find the whole western blot of <a href="#cancers-12-03221-f003" class="html-fig">Figure 3</a>B in <a href="#app1-cancers-12-03221" class="html-app">Figure S9</a> (<b>C</b>) and mRNA. Please find the whole western blot of <a href="#cancers-12-03221-f003" class="html-fig">Figure 3</a>C in <a href="#app1-cancers-12-03221" class="html-app">Figure S10</a> (<b>D</b>) levels in GC muscle of mice injected with C26 cells (C26) or vehicle (CT). Coomassie blue staining was used as loading control. (<b>E</b>) Spearman correlation between log2 transformed <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA levels in GC muscle and body weight loss (%). (<b>F</b>) Spearman correlation between log2 transformed <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA levels, and log2 transformed <span class="html-italic">Atrogin-1</span> mRNA in GC muscle (<span class="html-italic">n</span> =24). Data are reported as means ± SEM (<span class="html-italic">n</span> = 5–10/group). ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. CT.</p>
Full article ">Figure 4
<p>Marked production of acute phase reactants (APR) is observed in the skeletal muscle of mouse models of cancer-induced cachexia. (<b>A</b>) Immunohistochemical staining for Hp, Serpina3n, C3, and SAA1 in FFPE sections of GC muscle of C26 and CT male mice. Scale bar = 100 µm. Scale bar = 10 µm in zoom insets. Blue arrow shows nuclei located at the periphery of the cell. Red arrow shows capillaries. Bracket shows muscle fiber containing the protein of interest. (<b>B</b>) <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA relative levels in the GC muscle of female mice injected with BaF3 cells (BaF3) or vehicle (CT) and (<b>C</b>) <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA relative levels in the GC muscle of KP53 or CT male mice. Data are reported as means ± SEM (<span class="html-italic">n</span> = 5–10/group). * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 vs. CT.</p>
Full article ">Figure 5
<p>Muscular expression of APR is regulated by IL-6 in vivo and by glucocorticoids/pro inflammatory cytokines in vitro. (<b>A</b>) Anti-IL-6 antibodies prevent the increased muscular production of acute phase reactants (APR) in C26 male mice. <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA levels in the GC muscle of mice injected with C26 cells alone (C26 + PBS) or in combination with rat anti-murine IL-6 antibodies (C26 + anti-IL-6) or with rat IgG1 isotype control (C26 + IgG) or injected with vehicle alone (CT + PBS). (B-C) Glucocorticoids and proinflammatory cytokines stimulate the production of acute phase reactants (APR) by skeletal muscle cells. <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA levels in C<sub>2</sub>C<sub>12</sub> myotubes exposed for 48 h to (<b>B</b>) IL-6 (25 ng/mL), dexamethasone (Dexa; 10<sup>−6</sup> M) alone or in combination. (<b>C</b>) <span class="html-italic">Hp</span>, <span class="html-italic">Serpina3n</span>, <span class="html-italic">C3</span>, and <span class="html-italic">Saa1</span> mRNA and protein levels in C<sub>2</sub>C<sub>12</sub> myotubes exposed for 48 h to TNF-α (5 ng/ml), IFN-γ (5 ng/mL) alone or in combination. Coomassie blue staining was used as loading control. Data are reported as means ± SEM (<span class="html-italic">n</span> = 3–8/group). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. ns: no statistically significant when compared to CT. Please find the whole western blot of <a href="#cancers-12-03221-f005" class="html-fig">Figure 5</a>C in the <a href="#app1-cancers-12-03221" class="html-app">Figure S12</a>.</p>
Full article ">Figure 6
<p>Muscle expression of acute phase reactants (APR) is increased in cancer patients and is inversely correlated with muscle index and muscle density. (<b>A</b>) <span class="html-italic">HP</span>, <span class="html-italic">SerpinA3</span>, <span class="html-italic">C3</span>, and <span class="html-italic">SAA1</span> mRNA relative levels in quadriceps biopsies of healthy subjects (CT), non-cachectic (CNC), and cachectic (CC) cancer patients. (<b>B</b>) Heatmap showing the Pearson correlation coefficients between muscular expression of four selected acute phase reactants in CNC and CC patients (<span class="html-italic">n</span> = 35) and, respectively, weight loss (%), skeletal muscle index (cm<sup>2</sup>/m<sup>2</sup>), skeletal muscle density (HU), muscle strength corrected with values for the 50th percentile (kg/kg) and circulating levels of IL-6, CRP, and albumin. Circulating HP concentrations are increased in CC patients and are predictive of poor survival. (<b>C</b>) Circulating levels (g/L) of HP in CT subjects, CNC and CC patients. Data are reported as median ± 95% confidence interval (CT, <span class="html-italic">n</span> =  8; CNC, <span class="html-italic">n</span> = 19; CC, <span class="html-italic">n</span> = 15). (<b>D</b>) Kaplan–Meier survival curves according to plasma haptoglobin levels (two groups determined with normal values range of haptoglobin: 2 mg/mL) for lung cancer patients (<span class="html-italic">n</span> = 57). Significance was determined by logrank test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
Back to TopTop